1
|
Taylor SE, Behr S, Cooper KL, Mahdi H, Fabian D, Gallion H, Ueland F, Vargo J, Orr B, Girda E, Courtney-Brooks M, Olawaiye AB, Randall LM, Richardson DL, Sullivan SA, Huang M, Christner SM, Beriwal S, Lin Y, Chauhan A, Chu E, Kohn EC, Kunos C, Ivy SP, Beumer JH. Dose finding, bioavailability, and PK-PD of oral triapine with concurrent chemoradiation for locally advanced cervical cancer and vaginal cancer (ETCTN 9892). Cancer Chemother Pharmacol 2024; 95:4. [PMID: 39673591 DOI: 10.1007/s00280-024-04720-1] [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: 07/25/2024] [Accepted: 11/16/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND The addition of IV triapine to chemoradiation appeared active in phase I and II studies but drug delivery is cumbersome. We examined PO triapine with cisplatin chemoradiation. METHODS We implemented a 3 + 3 design for PO triapine dose escalation with expansion, starting at 100 mg, five days a week for five weeks while receiving radiation with weekly IV cisplatin for locally advanced cervical or vaginal cancer. Maximum tolerated dose (MTD), dose limiting toxicity (DLT), adverse events, pharmacokinetics (PK), pharmacodynamics (PD), and metabolic complete response (mCR) were assessed. RESULTS 19/21 patients were DLT evaluable. DLTs included grade 4 neutropenia (n = 2), leukopenia (n = 2), lymphopenia (n = 2), and hypokalemia (n = 1). Grade 3 toxicities at least possibly related were as expected for cisplatin chemoradiation: lymphopenia (n = 12), anemia (n = 10), neutropenia (n = 4), leukopenia (n = 8), decreased platelets (n = 2), hypertension (n = 1), and hyponatremia (n = 1). MTD and RP2D were established at 100 mg. 8/13 evaluable patients had a mCR. Triapine had a bioavailability of 59%. Methemoglobin levels correlated with triapine exposure. Smoking almost doubled CYP1A2 mediated triapine clearance. CONCLUSIONS Oral triapine is safe when given with cisplatin chemoradiation, convenient, bioavailable. Exposure is negatively impacted by smoking, and methemoglobin is a biomarker of exposure. CLINICAL TRIAL REGISTRATION NCT02595879.
Collapse
Affiliation(s)
- Sarah E Taylor
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| | - Sarah Behr
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Kristine L Cooper
- Biostatistics Facility, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haider Mahdi
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Cancer Immunology Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | | | | | | | - John Vargo
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Brian Orr
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eugenia Girda
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Madeleine Courtney-Brooks
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexander B Olawaiye
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Leslie M Randall
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology and Massey Comprehensive Cancer Center, Virginia Commonwealth University Health, Richmond, VA, USA
| | - Debra L Richardson
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stephanie A Sullivan
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology and Massey Comprehensive Cancer Center, Virginia Commonwealth University Health, Richmond, VA, USA
| | - Marilyn Huang
- Division of Gynecologic Oncology, University of Virginia, Charlottesville, VA, USA
| | - Susan M Christner
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Sushil Beriwal
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Yan Lin
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Biostatistics Facility, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aman Chauhan
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Edward Chu
- Montefiore Einstein Cancer Canter, Bronx, NY, USA
| | - Elise C Kohn
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Charles Kunos
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - S Percy Ivy
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Jan H Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, Pittsburgh, PA, USA.
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Kaya B, Smith H, Chen Y, Azad MG, Russell TM, Richardson V, Dharmasivam M, Richardson DR. Innovative N-Acridine Thiosemicarbazones and Their Zn(II) Complexes Transmetallate with Cu(II): Redox Activity and Suppression of Detrimental Oxy-Myoglobin Oxidation. Inorg Chem 2024; 63:20840-20858. [PMID: 39404641 DOI: 10.1021/acs.inorgchem.4c03642] [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: 10/29/2024]
Abstract
The coordination chemistry and electrochemistry of novel N-acridine thiosemicarbazones (NATs) were investigated along with their redox activity, antiproliferative efficacy, transmetalation, and dissociation properties. The ability of NAT Fe(III) complexes to inhibit detrimental oxy-myoglobin (oxy-Mb) oxidation was also examined. The NATs act as tridentate ligands with a 2:1 L/Zn(II) complex crystal structure, revealing a distorted octahedral geometry, where both ligands bind Zn(II) in a meridional conformation. The NAT Fe(III) complexes exhibited fully reversible one-electron FeIII/II couples with more positive potentials than the Fe(III) complexes of a related clinically trialed thiosemicarbazone (e.g., [Fe(DpC)2]+) due to the electron-donating capacity of acridine. Surprisingly, the NAT-Zn(II) complexes showed generally greater or similar antiproliferative activity than their ligands, Cu(II), or Fe(III) complexes. This may be explained by (1) formation of a highly lipophilic Zn(II) complex that acts as a chaperone to promote cellular uptake and (2) the capacity of the Zn(II) complex to dissociate or undergo transmetalation to the redox-active Cu(II) complex. Of the NAT-Fe(III) complexes, [Fe(AOBP)2]+ demonstrated a significant (p < 0.0001) improvement in preventing oxy-Mb oxidation than the Fe(III) complex of the clinically trialed thiosemicarbazone, DpC. This article advances our understanding of NAT coordination chemistry, electrochemistry, and the intriguing biological activity of their complexes.
Collapse
Affiliation(s)
- Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Henry Smith
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Yanbing Chen
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Tiffany M Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
3
|
Kaya B, Gholam Azad M, Suleymanoglu M, Harmer JR, Wijesinghe TP, Richardson V, Zhao X, Bernhardt PV, Dharmasivam M, Richardson DR. Isosteric Replacement of Sulfur to Selenium in a Thiosemicarbazone: Promotion of Zn(II) Complex Dissociation and Transmetalation to Augment Anticancer Efficacy. J Med Chem 2024; 67:12155-12183. [PMID: 38967641 DOI: 10.1021/acs.jmedchem.4c00884] [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: 07/06/2024]
Abstract
We implemented isosteric replacement of sulfur to selenium in a novel thiosemicarbazone (PPTP4c4mT) to create a selenosemicarbazone (PPTP4c4mSe) that demonstrates potentiated anticancer efficacy and selectivity. Their design specifically incorporated cyclohexyl and styryl moieties to sterically inhibit the approach of their Fe(III) complexes to the oxy-myoglobin heme plane. Importantly, in contrast to the Fe(III) complexes of the clinically trialed thiosemicarbazones Triapine, COTI-2, and DpC, the Fe(III) complexes of PPTP4c4mT and PPTP4c4mSe did not induce detrimental oxy-myoglobin oxidation. Furthermore, PPTP4c4mSe demonstrated more potent antiproliferative activity than the homologous thiosemicarbazone, PPTP4c4mT, with their selectivity being superior or similar, respectively, to the clinically trialed thiosemicarbazone, COTI-2. An advantageous property of the selenosemicarbazone Zn(II) complexes relative to their thiosemicarbazone analogues was their greater transmetalation to Cu(II) complexes in lysosomes. This latter effect probably promoted their antiproliferative activity. Both ligands down-regulated multiple key receptors that display inter-receptor cooperation that leads to aggressive and resistant breast cancer.
Collapse
Affiliation(s)
- Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Mediha Suleymanoglu
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Fatih, Istanbul 34093, Turkey
| | - Jeffrey R Harmer
- Centre for Advanced Imaging, University of Queensland, Brisbane 4072, Australia
| | - Tharushi P Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Xiao Zhao
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane 4072, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
4
|
Ueno H, Hoshino T, Yano W, Tsukioka S, Suzuki T, Hara S, Ogino Y, Chong KT, Suzuki T, Tsuji S, Itadani H, Yamamiya I, Otsu Y, Ito S, Yonekura T, Terasaka M, Tanaka N, Miyahara S. TAS1553, a small molecule subunit interaction inhibitor of ribonucleotide reductase, exhibits antitumor activity by causing DNA replication stress. Commun Biol 2022; 5:571. [PMID: 35681099 PMCID: PMC9184620 DOI: 10.1038/s42003-022-03516-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/22/2022] [Indexed: 01/03/2023] Open
Abstract
Ribonucleotide reductase (RNR) is composed of two non-identical subunits, R1 and R2, and plays a crucial role in balancing the cellular dNTP pool, establishing it as an attractive cancer target. Herein, we report the discovery of a highly potent and selective small-molecule inhibitor, TAS1553, targeting protein-protein interaction between R1 and R2. TAS1553 is also expected to demonstrate superior selectivity because it does not directly target free radical or a substrate binding site. TAS1553 has shown antiproliferative activity in human cancer cell lines, dramatically reducing the intracellular dATP pool and causing DNA replication stress. Furthermore, we identified SLFN11 as a biomarker that predicts the cytotoxic effect of TAS1553. Oral administration of TAS1553 demonstrated robust antitumor efficacy against both hematological and solid cancer xenograft tumors and also provided a significant survival benefit in an acute myelogenous leukemia model. Our findings strongly support the evaluation of TAS1553 in clinical trials. A small-molecule protein-protein interaction inhibitor of ribonucleotide reductase subunit, TAS1553, is shown to inhibit growth of both hematological and solid cancer xenograft tumors following oral administration in mice.
Collapse
|
5
|
Mathuber M, Hager S, Keppler BK, Heffeter P, Kowol CR. Liposomal formulations of anticancer copper(II) thiosemicarbazone complexes. Dalton Trans 2021; 50:16053-16066. [PMID: 34617075 PMCID: PMC8594434 DOI: 10.1039/d1dt02763h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/27/2021] [Indexed: 11/21/2022]
Abstract
α-N-Heterocyclic thiosemicarbazones such as triapine and COTI-2 are currently investigated as anticancer therapeutics in clinical trials. However, triapine was widely inactive against solid tumor types. A likely explanation is the short plasma half-life time and fast metabolism. One promising approach to overcome these drawbacks is the encapsulation of the drug into nanoparticles (passive drug-targeting). In a previous work we showed that it was not possible to stably encapsulate free triapine into liposomes. Hence, in this manuscript we present the successful preparation of liposomal formulations of the copper(II) complexes of triapine and COTI-2. To this end, various drug-loading strategies were examined and the resulting liposomes were physico-chemically characterized. Especially for liposomal Cu-triapine, a decent encapsulation efficacy and a slow drug release behavior could be observed. In contrast, for COTI-2 and its copper(II) complex no stable loading could be achieved. Subsequent in vitro studies in different cell lines with liposomal Cu-triapine showed the expected strongly reduced cytotoxicity and DNA damage induction. Also in vivo distinctly higher copper plasma levels and a continuous release could be observed for the liposomal formulation compared to free Cu-triapine. Taken together, the here presented nanoformulation of Cu-triapine is an important step further to increase the plasma half-life time and tumor targeting properties of anticancer thiosemicarbazones.
Collapse
Affiliation(s)
- Marlene Mathuber
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Straße 42, 1090 Vienna, Austria.
| | - Sonja Hager
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
- Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Straße 42, 1090 Vienna, Austria.
- Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Vienna, Austria
- Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Straße 42, 1090 Vienna, Austria.
- Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
6
|
Ohmura S, Marchetto A, Orth MF, Li J, Jabar S, Ranft A, Vinca E, Ceranski K, Carreño-Gonzalez MJ, Romero-Pérez L, Wehweck FS, Musa J, Bestvater F, Knott MML, Hölting TLB, Hartmann W, Dirksen U, Kirchner T, Cidre-Aranaz F, Grünewald TGP. Translational evidence for RRM2 as a prognostic biomarker and therapeutic target in Ewing sarcoma. Mol Cancer 2021; 20:97. [PMID: 34315482 PMCID: PMC8314608 DOI: 10.1186/s12943-021-01393-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 07/12/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Shunya Ohmura
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ) & Hopp-Children's Cancer Center (KiTZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany.,Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Aruna Marchetto
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Martin F Orth
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Jing Li
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ) & Hopp-Children's Cancer Center (KiTZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany
| | - Susanne Jabar
- Pediatrics III, West German Cancer Centre, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK), partner site Essen, Essen, Germany
| | - Andreas Ranft
- Pediatrics III, West German Cancer Centre, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK), partner site Essen, Essen, Germany
| | - Endrit Vinca
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ) & Hopp-Children's Cancer Center (KiTZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany
| | - Katharina Ceranski
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ) & Hopp-Children's Cancer Center (KiTZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany
| | - Martha J Carreño-Gonzalez
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ) & Hopp-Children's Cancer Center (KiTZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany
| | - Laura Romero-Pérez
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ) & Hopp-Children's Cancer Center (KiTZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany.,Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Fabienne S Wehweck
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Julian Musa
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ) & Hopp-Children's Cancer Center (KiTZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany.,Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Bestvater
- Light Microscopy Facility, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Maximilian M L Knott
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Tilman L B Hölting
- Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Wolfgang Hartmann
- Division of Translational Pathology, Gerhard-Domagk-Institute for Pathology, University Hospital Münster, Münster, Germany
| | - Uta Dirksen
- Pediatrics III, West German Cancer Centre, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK), partner site Essen, Essen, Germany
| | - Thomas Kirchner
- Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Florencia Cidre-Aranaz
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ) & Hopp-Children's Cancer Center (KiTZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany.,Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Thomas G P Grünewald
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany. .,Division of Translational Pediatric Sarcoma Research (B410), German Cancer Research Center (DKFZ) & Hopp-Children's Cancer Center (KiTZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany. .,Max-Eder Research Group for Pediatric Sarcoma Biology, Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany. .,Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
7
|
Lin ZP, Al Zouabi NN, Xu ML, Bowen NE, Wu TL, Lavi ES, Huang PH, Zhu YL, Kim B, Ratner ES. In silico screening identifies a novel small molecule inhibitor that counteracts PARP inhibitor resistance in ovarian cancer. Sci Rep 2021; 11:8042. [PMID: 33850183 PMCID: PMC8044145 DOI: 10.1038/s41598-021-87325-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
Poly ADP-ribose polymerase (PARP) inhibitors are promising targeted therapy for epithelial ovarian cancer (EOC) with BRCA mutations or defective homologous recombination (HR) repair. However, reversion of BRCA mutation and restoration of HR repair in EOC lead to PARP inhibitor resistance and reduced clinical efficacy of PARP inhibitors. We have previously shown that triapine, a small molecule inhibitor of ribonucleotide reductase (RNR), impaired HR repair and sensitized HR repair-proficient EOC to PARP inhibitors. In this study, we performed in silico screening of small molecule libraries to identify novel compounds that bind to the triapine-binding pocket on the R2 subunit of RNR and inhibit RNR in EOC cells. Following experimental validation of selected top-ranking in silico hits for inhibition of dNTP and DNA synthesis, we identified, DB4, a putative RNR pocket-binding inhibitor markedly abrogated HR repair and sensitized BRCA-wild-type EOC cells to the PARP inhibitor olaparib. Furthermore, we demonstrated that the combination of DB4 and olaparib deterred the progression of BRCA-wild type EOC xenografts and significantly prolonged the survival time of tumor-bearing mice. Herein we report the discovery of a putative small molecule inhibitor of RNR and HR repair for combination with PARP inhibitors to treat PARP inhibitor-resistant and HR repair-proficient EOC.
Collapse
Affiliation(s)
- Z Ping Lin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA.
| | - Nour N Al Zouabi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Mark L Xu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Nicole E Bowen
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Terence L Wu
- Yale West Campus Analytical Core, Yale University, West Haven, CT, 06516, USA
| | - Ethan S Lavi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Pamela H Huang
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Yong-Lian Zhu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Baek Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Drug Discovery, Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Elena S Ratner
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
8
|
Selenotriapine – An isostere of the most studied thiosemicarbazone with pronounced pro-apoptotic activity, low toxicity and ability to challenge phenotype reprogramming of 3-D mammary adenocarcinoma tumors. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2017.11.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
9
|
Kunos CA, Andrews SJ, Moore KN, Chon HS, Ivy SP. Randomized Phase II Trial of Triapine-Cisplatin-Radiotherapy for Locally Advanced Stage Uterine Cervix or Vaginal Cancers. Front Oncol 2019; 9:1067. [PMID: 31681600 PMCID: PMC6803528 DOI: 10.3389/fonc.2019.01067] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/30/2019] [Indexed: 11/13/2022] Open
Abstract
Uterine cervix or vaginal cancers have inherent overactivity of ribonucleotide reductase (RNR), making these cancers rational targets for therapy based on interruption of cisplatin-radiotherapy-induced DNA damage repair. We conducted a pilot, open-label randomized phase II trial to evaluate the efficacy and safety of cisplatin-radiotherapy with or without triapine, a small molecule with RNR-inhibitory activity, in patients with advanced-stage uterine cervix or vaginal cancers (NCT01835171), as a lead in to a randomized phase III study (NCT02466971). A total of 26 women were randomly assigned to receive 6 weeks of daily radiotherapy followed by brachytherapy (80 Gy) and once-weekly cisplatin (40 mg m−2)—with or without three-times weekly intravenous triapine (25 mg m−2)—in one 56-days cycle. Primary end points were metabolic complete response by positron emission tomography and safety. Additional end points included the rate of clinical response, rate of methemoglobinemia, and progression-free survival. The addition of triapine to cisplatin-radiotherapy improved the rate of metabolic complete response from 69 to 92% (P = 0.32) and raised the 3-year progression-free survival estimate from 77 to 92% (hazard ratio for progression, 0.30; P = 0.27). The most frequent grade 3 or 4 adverse events in either treatment group included reversible leukopenia, neutropenia, fatigue, or electrolyte abnormalities. No significant differences were seen between the two groups in the rate of adverse events. Symptomatic methemoglobinemia was not encountered after triapine infusion. In conclusion, the addition of triapine to cisplatin-radiotherapy improved the rate of metabolic complete response in patients with advanced-stage uterine cervix or vaginal cancers without significant toxicity. A phase III trial adequately powered to evaluate progression-free and overall survival is underway (NCT02466971).
Collapse
Affiliation(s)
- Charles A Kunos
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, United States
| | | | - Kathleen N Moore
- University of Oklahoma Stephenson Cancer Center, Oklahoma City, OK, United States
| | - Hye Sook Chon
- H. Lee Moffitt Cancer & Research Institute, Tampa, FL, United States
| | - S Percy Ivy
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
10
|
Ivy SP, Kunos CA, Arnaldez FI, Kohn EC. Defining and targeting wild-type BRCA high-grade serous ovarian cancer: DNA repair and cell cycle checkpoints. Expert Opin Investig Drugs 2019; 28:771-785. [PMID: 31449760 DOI: 10.1080/13543784.2019.1657403] [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] [Indexed: 12/18/2022]
Abstract
Introduction: Molecular analyzes including molecular descriptor/phenotype interactions have led to better characterization of epithelial ovarian cancer patients, including a definition of a BRCA wild-type (BRCAwt) phenotype. Understanding how and when to use agents targeted against dependent BRCAwt pathways or other molecular events at disease progression is an important translational and therapeutic direction in ovarian cancer research. Areas covered: In this overview, we provide definitions and descriptions of a BRCAwt genotype and phenotype. We discuss novel investigational drugs that hold promise for the treatment of BRCAwt ovarian cancer, including inhibitors of poly(ADP-ribose) polymerase, ribonucleotide reductase, DNA protein kinase-catalytic subunit, ataxia-telangiectasia-mutated kinase (ATM), ataxia-telangiectasia mutated and Rad3-related kinase (ATR), CHK 1/2, cyclin kinases, glutaminase-1, WEE1 kinase, as well as tumor microenvironment and angiogenesis inhibitors. This article explores the known and the emerging areas of clinical research on patients with BRCAwt ovarian cancer. Expert opinion: Discovery of molecular changes tied to annotated disease information, along with an expanding array of pathway targets and targeted therapeutic agents, creates optimism and opportunity for women with ovarian cancer. Using precision oncology approaches, clinical researchers are, and will be, poised to select more effective treatments for ovarian cancer patients.
Collapse
Affiliation(s)
- S Percy Ivy
- Cancer Therapy Evaluation Program, National Cancer Institute (NCI) , Bethesda , MD , USA
| | - Charles A Kunos
- Cancer Therapy Evaluation Program, National Cancer Institute (NCI) , Bethesda , MD , USA
| | - Fernanda I Arnaldez
- Cancer Therapy Evaluation Program, National Cancer Institute (NCI) , Bethesda , MD , USA
| | - Elise C Kohn
- Cancer Therapy Evaluation Program, National Cancer Institute (NCI) , Bethesda , MD , USA
| |
Collapse
|
11
|
Heffeter P, Pape VFS, Enyedy ÉA, Keppler BK, Szakacs G, Kowol CR. Anticancer Thiosemicarbazones: Chemical Properties, Interaction with Iron Metabolism, and Resistance Development. Antioxid Redox Signal 2019; 30:1062-1082. [PMID: 29334758 DOI: 10.1089/ars.2017.7487] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE During the past decades, thiosemicarbazones were clinically developed for a variety of diseases, including tuberculosis, viral infections, malaria, and cancer. With regard to malignant diseases, the class of α-N-heterocyclic thiosemicarbazones, and here especially 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (Triapine), was intensively developed in multiple clinical phase I/II trials. Recent Advances: Very recently, two new derivatives, namely COTI-2 and di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) have entered phase I evaluation. Based on the strong metal-chelating/metal-interacting properties of thiosemicarbazones, interference with the cellular iron (and copper) homeostasis is assumed to play an important role in their biological activity. CRITICAL ISSUES In this review, we summarize and analyze the data on the interaction of (α-N-heterocyclic) thiosemicarbazones with iron, with the special aim of bridging the current knowledge on their mode of action from chemistry to (cell) biology. In addition, we highlight the difference to classical iron(III) chelators such as desferrioxamine (DFO), which are used for the treatment of iron overload. FUTURE DIRECTIONS We want to emphasize that thiosemicarbazones are not solely removing iron from the cells/organism. In contrast, they should be considered as iron-interacting drugs influencing diverse biological pathways in a complex and multi-faceted mode of action. Consequently, in addition to the discussion of physicochemical properties (e.g., complex stability, redox activity), this review contains an overview on the diversity of cellular thiosemicarbazone targets and drug resistance mechanisms.
Collapse
Affiliation(s)
- Petra Heffeter
- 1 Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna , Vienna, Austria .,2 Research Cluster "Translational Cancer Therapy Research," Vienna, Austria
| | - Veronika F S Pape
- 3 Institute of Enzymology , Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary .,4 Department of Physiology, Faculty of Medicine, Semmelweis University , Budapest, Hungary
| | - Éva A Enyedy
- 5 Department of Inorganic and Analytical Chemistry, University of Szeged , Szeged, Hungary
| | - Bernhard K Keppler
- 2 Research Cluster "Translational Cancer Therapy Research," Vienna, Austria .,6 Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna , Vienna, Austria
| | - Gergely Szakacs
- 1 Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center of the Medical University, Medical University of Vienna , Vienna, Austria .,3 Institute of Enzymology , Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Christian R Kowol
- 2 Research Cluster "Translational Cancer Therapy Research," Vienna, Austria .,6 Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna , Vienna, Austria
| |
Collapse
|
12
|
Darcy H, Simpson K, Gajanayake I, Seth M, McGrotty Y, Szladovits B, Glanemann B. Feline primary erythrocytosis: a multicentre case series of 18 cats. J Feline Med Surg 2018; 20:1192-1198. [PMID: 29364032 PMCID: PMC11104208 DOI: 10.1177/1098612x17750333] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CASE SERIES SUMMARY A retrospective multicentre case series of feline primary erythrocytosis (PE) was evaluated. The aim was to gain better understanding of disease presentation and progression to guide management and prognostication. Case records were assessed for evidence of increased packed cell volume (PCV; >48%), sufficient investigation to rule out relative and secondary erythrocytosis, and follow-up data for at least 12 months or until death. Eighteen cats were included in the case series. No significant trends in signalment were noted. Seizures and mentation changes were the most common presenting signs (both n = 10). Median PCV was 70% (median total protein concentration of 76 g/l) with no other consistent haematological changes. Sixteen cats survived to discharge. Phlebotomy was performed initially in 15/16 surviving animals and performed after discharge in 10/16. Hydroxyurea was the most common adjunctive therapy, used in 10/16 cats. Of the 16 patients surviving to discharge, 14 patients were still alive at the conclusion of the study (survival time >17 months post-discharge), with the two non-survivors having lived for 5 years or more after diagnosis. PCV, when stabilised, did not correlate with resolution of clinical signs. RELEVANCE AND NOVEL INFORMATION In contrast to perceptions, feline PE was generally well managed via a combination of phlebotomy and medical therapy, with evidence of prolonged survival times. The use of hydroxyurea enabled cessation or repeat phlebotomies.
Collapse
Affiliation(s)
- Hannah Darcy
- Queen Mother Hospital for Animals, Department of Clinical Science and Services, Royal Veterinary College, North Mymms, Hertfordshire, UK
| | - Katherine Simpson
- Goddard Veterinary Group, Mandeville Veterinary Hospital, Northolt, UK
| | - Isuru Gajanayake
- Willows Veterinary Centre and Referral Service, Solihull, West Midlands, UK
| | - Mayank Seth
- Centre for Small Animal Studies, Animal Health Trust, Newmarket, Suffolk, UK
| | | | - Balazs Szladovits
- Diagnostic Laboratory Services, Department of Pathobiology and Population Sciences, Royal Veterinary College, North Mymms, Hertfordshire, UK
| | - Barbara Glanemann
- Queen Mother Hospital for Animals, Department of Clinical Science and Services, Royal Veterinary College, North Mymms, Hertfordshire, UK
| |
Collapse
|
13
|
Lin ZP, Zhu YL, Lo YC, Moscarelli J, Xiong A, Korayem Y, Huang PH, Giri S, LoRusso P, Ratner ES. Combination of triapine, olaparib, and cediranib suppresses progression of BRCA-wild type and PARP inhibitor-resistant epithelial ovarian cancer. PLoS One 2018; 13:e0207399. [PMID: 30444904 PMCID: PMC6239325 DOI: 10.1371/journal.pone.0207399] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/30/2018] [Indexed: 12/14/2022] Open
Abstract
PARP inhibitors target BRCA mutations and defective homologous recombination repair (HRR) for the treatment of epithelial ovarian cancer (EOC). However, the treatment of HRR-proficient EOC with PARP inhibitors remains challenging. The objective of this study was to determine whether the combination of triapine (ribonucleotide reductase inhibitor), cediranib (vascular endothelial growth factor receptor tyrosine kinase inhibitor), and the PARP inhibitor olaparib synergized against BRCA wild-type and HRR-proficient EOC in xenograft mouse models. In addition, the mechanisms by which cediranib augmented the efficacy of triapine and olaparib were investigated. BRCA-wild type and PARP inhibitor-resistant EOC cell lines were implanted subcutaneously (s.c.) into nude mice or injected intraperitoneally (i.p.) into SCID-Beige mice. Mice were then treated i.p. with olaparib, cediranib, triapine, various double and triple combinations. The volume of s.c tumor in nude mice and the abdominal circumference of SCID-Beige mice were measured to evaluate the effectiveness of the treatment to delay tumor growth and prolong the survival time of mice. In both xenograft mouse models, the combination of triapine, olaparib and cediranib resulted in marked suppression of BRCA-wild type EOC growth and significant prolongation of the survival time of mice, with efficacy greater than any double combinations and single drugs. Furthermore, we identified that cediranib abrogated pro-survival and anti-apoptotic AKT signaling, thereby enhancing the efficacy of triapine and olaparib against BRCA-wild type EOC cells. Taken together, our results demonstrate a proof-of-principle approach and the combination regiment holds promise in treating BRCA-wild type and PARP inhibitor-resistant EOC.
Collapse
Affiliation(s)
- Z. Ping Lin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail: (ESR); (ZPL)
| | - Yong-Lian Zhu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Ying-Chun Lo
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jake Moscarelli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Amy Xiong
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Yasmin Korayem
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Pamela H. Huang
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Smith Giri
- Section of Medical Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Patricia LoRusso
- Section of Medical Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Elena S. Ratner
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, United States of America
- * E-mail: (ESR); (ZPL)
| |
Collapse
|
14
|
Lin ZP, Zhu YL, Ratner ES. Targeting Cyclin-Dependent Kinases for Treatment of Gynecologic Cancers. Front Oncol 2018; 8:303. [PMID: 30135856 PMCID: PMC6092490 DOI: 10.3389/fonc.2018.00303] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/19/2018] [Indexed: 02/01/2023] Open
Abstract
Ovarian, uterine/endometrial, and cervical cancers are major gynecologic malignancies estimated to cause nearly 30,000 deaths in 2018 in US. Defective cell cycle regulation is the hallmark of cancers underpinning the development and progression of the disease. Normal cell cycle is driven by the coordinated and sequential rise and fall of cyclin-dependent kinases (CDK) activity. The transition of cell cycle phases is governed by the respective checkpoints that prevent the entry into the next phase until cellular or genetic defects are repaired. Checkpoint activation is achieved by p53- and ATM/ATR-mediated inactivation of CDKs in response to DNA damage. Therefore, an aberrant increase in CDK activity and/or defects in checkpoint activation lead to unrestricted cell cycle phase transition and uncontrolled proliferation that give rise to cancers and perpetuate malignant progression. Given that CDK activity is also required for homologous recombination (HR) repair, pharmacological inhibition of CDKs can be exploited as a synthetic lethal approach to augment the therapeutic efficacy of PARP inhibitors and other DNA damaging modalities for the treatment of gynecologic cancers. Here, we overview the basic of cell cycle and discuss the mechanistic studies that establish the intimate link between CDKs and HR repair. In addition, we present the perspective of preclinical and clinical development in small molecule inhibitors of CDKs and CDK-associated protein targets, as well as their potential use in combination with hormonal therapy, PARP inhibitors, chemotherapy, and radiation to improve treatment outcomes.
Collapse
Affiliation(s)
- Z Ping Lin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, United States
| | - Yong-Lian Zhu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, United States
| | - Elena S Ratner
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
15
|
Kunos CA, Ivy SP. Triapine Radiochemotherapy in Advanced Stage Cervical Cancer. Front Oncol 2018; 8:149. [PMID: 29868473 PMCID: PMC5949312 DOI: 10.3389/fonc.2018.00149] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/23/2018] [Indexed: 12/23/2022] Open
Abstract
Clinical ribonucleotide reductase (RNR) inhibitors have reinvigorated enthusiasm for radiochemotherapy treatment of patients with regionally advanced stage cervical cancers. About two-thirds of patients outlive their cervical cancer (1), even though up to half of their tumors retain residual microscopic disease (2). The National Cancer Institute Cancer Therapy Evaluation Program conducted two prospective trials of triapine–cisplatin–radiation to improve upon this finding by precisely targeting cervical cancer’s overactive RNR. Triapine’s potent inactivation of RNR arrests cells at the G1/S cell cycle restriction checkpoint and enhances cisplatin–radiation cytotoxicity. In this article, we provide perspective on challenges encountered in and future potential of clinical development of a triapine–cisplatin–radiation combination for patients with regionally advanced cervical cancer. New trial results and review presented here suggest that a triapine–cisplatin–radiation combination may offer molecular cell cycle target control to maximize damage in cancers and to minimize injury to normal cells. A randomized trial now accrues patients with regionally advanced stage cervical cancer to evaluate triapine’s contribution to clinical benefit after cisplatin–radiation (clinicaltrials.gov, NCT02466971).
Collapse
Affiliation(s)
- Charles A Kunos
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, United States
| | - S Percy Ivy
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
16
|
Kunos CA, Chu E, Beumer JH, Sznol M, Ivy SP. Phase I trial of daily triapine in combination with cisplatin chemotherapy for advanced-stage malignancies. Cancer Chemother Pharmacol 2016; 79:201-207. [PMID: 27878356 DOI: 10.1007/s00280-016-3200-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/11/2016] [Indexed: 11/29/2022]
Abstract
PURPOSE Advanced-stage malignancies have increased deoxyribonucleotide demands in DNA replication and repair, making deoxyribonucleotide supply a potential exploitable target for therapy based on ribonucleotide reductase (RNR) inhibition. METHODS A dose-finding phase I trial was conducted of intravenous (i.v.) triapine, a small-molecule RNR inhibitor, and cisplatin chemotherapy in patients with advanced-stage solid tumor malignancies. Patients received dose-finding levels of i.v. triapine (48-96 mg/m2) and i.v. cisplatin (20-75 mg/m2) on 1 of 3 different schedules. The primary endpoint was to identify the maximum tolerated dose of a triapine-cisplatin combination. Secondary endpoints included the rate of triapine-cisplatin objective response and the pharmacokinetics and bioavailability of a single oral triapine dose. (Clinicaltrials.gov number, NCT00024323). RESULTS The MTD was 96 mg/m2 triapine daily days 1-4 and 75 mg/m2 cisplatin split over day 2 and day 3. Frequent grade 3 or 4 adverse events included fatigue, dyspnea, leukopenia, thrombocytopenia, and electrolyte abnormalities. No objective responses were observed; 5 (50%) of 10 patients treated at the MTD had stable disease. Pharmacokinetics indicated an oral triapine bioavailability of 88%. CONCLUSIONS The triapine-cisplatin combination may be given safely in patients with advanced-stage solid tumor malignancies. On the basis of these results, a phase I trial adequately powered to evaluate oral triapine bioavailability in women with advanced-stage uterine cervix or vulvar cancers is underway.
Collapse
Affiliation(s)
- Charles A Kunos
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA.
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, MSC 9739, Rockville, MD, 20892-9760, USA.
| | - Edward Chu
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jan H Beumer
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
- University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Mario Sznol
- Yale University School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | - S Percy Ivy
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
17
|
Takebe N, Ahmed MM, Vikram B, Bernhard EJ, Zwiebel J, Norman Coleman C, Kunos CA. Radiation-Therapeutic Agent Clinical Trials: Leveraging Advantages of a National Cancer Institute Programmatic Collaboration. Semin Radiat Oncol 2016; 26:271-80. [PMID: 27619249 DOI: 10.1016/j.semradonc.2016.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A number of oncology phase II radiochemotherapy trials with promising results have been conducted late in the overall experimental therapeutic agent development process. Accelerated development and approval of experimental therapeutic agents have stimulated further interest in much earlier radiation-agent studies to increase the likelihood of success in phase III trials. To sustain this interest, more forward-thinking preclinical radiobiology experimental designs are needed to improve discovery of promising radiochemotherapy plus agent combinations for clinical trial testing. These experimental designs should better inform next-step radiation-agent clinical trial dose, schedule, exposure, and therapeutic effect. Recognizing the need for a better strategy to develop preclinical data supporting radiation-agent phase I or II trials, the National Cancer Institute (NCI)-Cancer Therapy Evaluation Program (CTEP) and the NCI-Molecular Radiation Therapeutics Branch of the Radiation Research Program have partnered to promote earlier radiobiology studies of CTEP portfolio agents. In this Seminars in Radiation Oncology article, four key components of this effort are discussed. First, we outline steps for accessing CTEP agents for preclinical testing. Second, we propose radiobiology studies that facilitate transition from preclinical testing to early phase trial activation. Third, we navigate steps that walk through CTEP agent strategic development paths available for radiation-agent testing. Fourth, we highlight a new NCI-sponsored cooperative agreement grant supporting in vitro and in vivo radiation-CTEP agent testing that informs early phase trial designs. Throughout the article, we include contemporary examples of successful radiation-agent development initiatives.
Collapse
Affiliation(s)
- Naoko Takebe
- Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| | - Mansoor M Ahmed
- Radiation Research Program, National Cancer Institute, Bethesda, MD
| | | | - Eric J Bernhard
- Radiation Research Program, National Cancer Institute, Bethesda, MD
| | - James Zwiebel
- Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - C Norman Coleman
- Radiation Research Program, National Cancer Institute, Bethesda, MD
| | - Charles A Kunos
- Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
18
|
Fischer B, Kryeziu K, Kallus S, Heffeter P, Berger W, Kowol CR, Keppler BK. Nanoformulations of anticancer thiosemicarbazones to reduce methemoglobin formation and improve anticancer activity. RSC Adv 2016. [DOI: 10.1039/c6ra07659a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Triapine and two derivatives were encapsulated into polymeric nanoparticles as well as liposomes. The most stable formulation showed strongly reduced methemoglobin formation and improved anticancer activity.
Collapse
Affiliation(s)
- Britta Fischer
- Institute of Inorganic Chemistry
- University of Vienna
- 1090 Vienna
- Austria
| | - Kushtrim Kryeziu
- Institute of Cancer Research and Comprehensive Cancer Center
- Medical University Vienna
- 1090 Vienna
- Austria
| | - Sebastian Kallus
- Institute of Inorganic Chemistry
- University of Vienna
- 1090 Vienna
- Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center
- Medical University Vienna
- 1090 Vienna
- Austria
- Research Platform “Translational Cancer Therapy Research”
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center
- Medical University Vienna
- 1090 Vienna
- Austria
- Research Platform “Translational Cancer Therapy Research”
| | - Christian R. Kowol
- Institute of Inorganic Chemistry
- University of Vienna
- 1090 Vienna
- Austria
- Research Platform “Translational Cancer Therapy Research”
| | - Bernhard K. Keppler
- Institute of Inorganic Chemistry
- University of Vienna
- 1090 Vienna
- Austria
- Research Platform “Translational Cancer Therapy Research”
| |
Collapse
|
19
|
Pelivan K, Miklos W, van Schoonhoven S, Koellensperger G, Gille L, Berger W, Heffeter P, Kowol CR, Keppler BK. Differences in protein binding and excretion of Triapine and its Fe(III) complex. J Inorg Biochem 2015; 160:61-9. [PMID: 26507768 DOI: 10.1016/j.jinorgbio.2015.10.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/02/2015] [Accepted: 10/05/2015] [Indexed: 11/26/2022]
Abstract
Triapine has been investigated as anticancer drug in multiple clinical phase I/II trials. Although promising anti-leukemic activity was observed, Triapine was ineffective against solid tumors. The reasons are currently widely unknown. The biological activity of Triapine is strongly connected to its iron complex (Fe-Triapine) which is pharmacologically not investigated. Here, novel analytical tools for Triapine and Fe-Triapine were developed and applied for cell extracts and body fluids of treated mice. Triapine and its iron complex showed a completely different behavior: for Triapine, low protein binding was observed in contrast to fast protein adduct formation of Fe-Triapine. Notably, both drugs were rapidly cleared from the body (serum half-life time <1h). Remarkably, in contrast to Triapine, where (in accordance to clinical data) basically no renal excretion was found, the iron complex was effectively excreted via urine. Moreover, no Fe-Triapine was detected in serum or cytosolic extracts after Triapine treatment. Taken together, our study will help to further understand the biological behavior of Triapine and its Fe-complex and allow the development of novel thiosemicarbazones with pronounced activity against solid tumor types.
Collapse
Affiliation(s)
- Karla Pelivan
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Strasse 42, 1090 Vienna, Austria
| | - Walter Miklos
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Sushilla van Schoonhoven
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Gunda Koellensperger
- Institute of Analytical Chemistry, University of Vienna, Waehringer Strasse 38, 1090 Vienna, Austria
| | - Lars Gille
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Vienna, Austria.
| | - Christian R Kowol
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Strasse 42, 1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Vienna, Austria.
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Strasse 42, 1090 Vienna, Austria; Research Platform "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Dutta P, Mondal S, Roy S, Lopez-Torres E, Sinha C. The structural characterization and DNA binding of pyridyl-azo-acetylacetonato complexes of zinc(II), cadmium(II) and mercury(II). Polyhedron 2015. [DOI: 10.1016/j.poly.2015.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
21
|
Aird KM, Zhang R. Nucleotide metabolism, oncogene-induced senescence and cancer. Cancer Lett 2014; 356:204-10. [PMID: 24486217 DOI: 10.1016/j.canlet.2014.01.017] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/06/2014] [Accepted: 01/22/2014] [Indexed: 01/28/2023]
Abstract
Senescence is defined as a stable cell growth arrest. Oncogene-induced senescence (OIS) occurs when an activated oncogene is expressed in a normal cell. OIS acts as a bona fide tumor suppressor mechanism by driving stable growth arrest of cancer progenitor cells harboring the initial oncogenic hit. OIS is often characterized by aberrant DNA replication and the associated DNA damage response. Nucleotides, in particular deoxyribonucleotide triphosphates (dNTPs), are necessary for both DNA replication and repair. Imbalanced dNTP pools play a role in a number of human diseases, including during the early stages of cancer development. This review will highlight what is currently known about the role of decreased nucleotide metabolism in OIS, how nucleotide metabolism leads to transformation and tumor progression, and how this pathway can be targeted as a cancer therapeutic by inducing senescence of cancer cells.
Collapse
Affiliation(s)
- Katherine M Aird
- Gene Expression and Regulation Program, The Wistar Institute Cancer Center, The Wistar Institute, Philadelphia, PA 19104, United States
| | - Rugang Zhang
- Gene Expression and Regulation Program, The Wistar Institute Cancer Center, The Wistar Institute, Philadelphia, PA 19104, United States.
| |
Collapse
|
22
|
Basha MT, Rodríguez C, Richardson DR, Martínez M, Bernhardt PV. Kinetic studies on the oxidation of oxyhemoglobin by biologically active iron thiosemicarbazone complexes: relevance to iron-chelator-induced methemoglobinemia. J Biol Inorg Chem 2013; 19:349-57. [PMID: 24317633 DOI: 10.1007/s00775-013-1070-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 11/18/2013] [Indexed: 12/29/2022]
Abstract
The oxidation of oxyhemoglobin to methemoglobin has been found to be facilitated by low molecular weight iron(III) thiosemicarbazone complexes. This deleterious reaction, which produces hemoglobin protein units unable to bind dioxygen and occurs during the administration of iron chelators such as the well-known 3-aminopyridine-2-pyridinecarbaldehyde thiosemicarbazone (3-AP; Triapine), has been observed in the reaction with Fe(III) complexes of some members of the 3-AP structurally-related thiosemicarbazone ligands derived from di-2-pyridyl ketone (HDpxxT series). We have studied the kinetics of this oxidation reaction in vitro using human hemoglobin and found that the reaction proceeds with two distinct time-resolved steps. These have been associated with sequential oxidation of the two different oxyheme cofactors in the α and β protein chains. Unexpected steric and hydrogen-bonding effects on the Fe(III) complexes appear to be the responsible for the observed differences in the reaction rate across the series of HDpxxT ligand complexes used in this study.
Collapse
Affiliation(s)
- Maram T Basha
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | | | | | | | | |
Collapse
|
23
|
Lee CH, Kim SH, Kwon DH, Jang KH, Chung YH, Moon JD. Two cases of methemoglobinemia induced by the exposure to nitrobenzene and aniline. Ann Occup Environ Med 2013; 25:31. [PMID: 24475977 PMCID: PMC3916109 DOI: 10.1186/2052-4374-25-31] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 10/18/2013] [Indexed: 11/10/2022] Open
Abstract
Objective To report two cases of methemoglobinemia induced by inhaled nitrobenzene and dermally absorbed aniline. Methods We have evaluated a 37-year-old male worker exposed to nitrobenzene by inhalation while conducting maintenance job of mononitrobenzene pump and a 25-year-old male worker exposed dermally to aniline while unloading. Results The first case is a 37-year-old male exposed to nitrobenzene. His blood methemoglobin concentration level was initially 19.8%, and chest X-ray was normal. After oxygen therapy, the blood methemoglobin concentration level decreased to 2.1%, and the symptoms were alleviated. The second case is a 25-year-old male exposed dermally to aniline. His chest X-ray was normal, but blood methemoglobin concentration level reached maximally 46.8%. He was treated with methylene blue due to relatively high blood methemoglobin level. Gradually after the treatment, his methemoglobin concentration level was normalized to 0.8% and simultaneously symptoms were resolved. Conclusions After the thorough exposure investigations and medical evaluations, we have concluded that these cases were methemoglobinemia induced by occupational exposure to nitrobenzene and aniline. We suggest that businesses which handle methemoglobinemia-causing substances control the engineering process strictly, implement periodic screening, and establish emergency patient management system.
Collapse
Affiliation(s)
| | | | | | | | | | - Jai Dong Moon
- Department of Occupational and Environmental Medicine, Chonnam National University Hwasun Hospital, Hwasun, Korea.
| |
Collapse
|
24
|
Kunos CA, Radivoyevitch T, Waggoner S, Debernardo R, Zanotti K, Resnick K, Fusco N, Adams R, Redline R, Faulhaber P, Dowlati A. Radiochemotherapy plus 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP, NSC #663249) in advanced-stage cervical and vaginal cancers. Gynecol Oncol 2013; 130:75-80. [PMID: 23603372 PMCID: PMC4260802 DOI: 10.1016/j.ygyno.2013.04.019] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 04/09/2013] [Accepted: 04/12/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Cervical and vaginal cancers have virally-mediated or mutated defects in DNA damage repair responses, making these cancers sensible targets for ribonucleotide reductase inhibition during radiochemotherapy. METHODS We conducted a phase II study evaluating 3× weekly 2-hour intravenous 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP, 25 mg/m(2)) co-administered with 1× weekly intravenous cisplatin (40 mg/m(2)) and daily pelvic radiation (45 Gy) in women with stage I(B2)-IV(B) cervical (n=22) or stage II-IV vaginal (n=3) cancers. Brachytherapy followed (40 Gy). Toxicity was monitored by common terminology criteria for adverse events (version 3.0). The primary end point of response was assessed by 3-month posttherapy 2-[(18)F] fluoro-2-deoxy-d-glucose positron emission tomography (PET/CT) and clinical examination. RESULTS 3-AP radiochemotherapy achieved clinical responses in 24 (96% [95% confidence interval: 80-99%]) of 25 patients (median follow-up 20 months, range 2-35 months). 23 (96% [95% confidence interval: 80-99%]) of 24 patients had 3-month posttherapy PET/CT scans that recorded metabolic activity in the cervix or vagina equal or less than that of the cardiac blood pool, suggesting complete metabolic responses. The most frequent 3-AP radiochemotherapy-related adverse events included fatigue, nausea, diarrhea, and reversible hematological and electrolyte abnormalities. CONCLUSIONS The addition of 3-AP to cisplatin radiochemotherapy was tolerable and produced high rates of clinical and metabolic responses in women with cervical and vaginal cancers. Future randomized phase II and III clinical trials of 3-AP radiochemotherapy are warranted.
Collapse
Affiliation(s)
- Charles A Kunos
- Department of Radiation Oncology, CASE Comprehensive Cancer Center, University Hospitals Case Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|