1
|
Zafeiropoulou K, Kalampounias G, Alexis S, Anastasopoulos D, Symeonidis A, Katsoris P. Autophagy and oxidative stress modulation mediate Bortezomib resistance in prostate cancer. PLoS One 2024; 19:e0289904. [PMID: 38412186 PMCID: PMC10898778 DOI: 10.1371/journal.pone.0289904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 01/27/2024] [Indexed: 02/29/2024] Open
Abstract
Proteasome inhibitors such as Bortezomib represent an established type of targeted treatment for several types of hematological malignancies, including multiple myeloma, Waldenstrom's macroglobulinemia, and mantle cell lymphoma, based on the cancer cell's susceptibility to impairment of the proteasome-ubiquitin system. However, a major problem limiting their efficacy is the emergence of resistance. Their application to solid tumors is currently being studied, while simultaneously, a wide spectrum of hematological cancers, such as Myelodysplastic Syndromes show minimal or no response to Bortezomib treatment. In this study, we utilize the prostate cancer cell line DU-145 to establish a model of Bortezomib resistance, studying the underlying mechanisms. Evaluating the resulting resistant cell line, we observed restoration of proteasome chymotrypsin-like activity, regardless of drug presence, an induction of pro-survival pathways, and the substitution of the Ubiquitin-Proteasome System role in proteostasis by induction of autophagy. Finally, an estimation of the oxidative condition of the cells indicated that the resistant clones reduce the generation of reactive oxygen species induced by Bortezomib to levels even lower than those induced in non-resistant cells. Our findings highlight the role of autophagy and oxidative stress regulation in Bortezomib resistance and elucidate key proteins of signaling pathways as potential pharmaceutical targets, which could increase the efficiency of proteasome-targeting therapies, thus expanding the group of molecular targets for neoplastic disorders.
Collapse
Affiliation(s)
- Kalliopi Zafeiropoulou
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
- Hematology Division, Department of Internal Medicine, University of Patras Medical School-University Hospital, Patras, Greece
| | - Georgios Kalampounias
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
| | - Spyridon Alexis
- Hematology Division, Department of Internal Medicine, University of Patras Medical School-University Hospital, Patras, Greece
| | - Daniil Anastasopoulos
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
| | - Argiris Symeonidis
- Hematology Division, Department of Internal Medicine, University of Patras Medical School-University Hospital, Patras, Greece
| | - Panagiotis Katsoris
- Division of Genetics, Cell Biology and Development, Department of Biology, University of Patras, Patras, Greece
| |
Collapse
|
2
|
Sprague JM, Yekkirala AS, Singh B, Tochitsky I, Stephens M, Viramontes O, Ivanis J, Biscola NP, Havton LA, Woolf CJ, Latremoliere A. Bortezomib-induced neuropathy is in part mediated by the sensitization of TRPV1 channels. Commun Biol 2023; 6:1228. [PMID: 38052846 PMCID: PMC10698173 DOI: 10.1038/s42003-023-05624-1] [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: 07/14/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023] Open
Abstract
TRPV1 is an ion channel that transduces noxious heat and chemical stimuli and is expressed in small fiber primary sensory neurons that represent almost half of skin nerve terminals. Tissue injury and inflammation result in the sensitization of TRPV1 and sustained activation of TRPV1 can lead to cellular toxicity though calcium influx. To identify signals that trigger TRPV1 sensitization after a 24-h exposure, we developed a phenotypic assay in mouse primary sensory neurons and performed an unbiased screen with a compound library of 480 diverse bioactive compounds. Chemotherapeutic agents, calcium ion deregulators and protein synthesis inhibitors were long-acting TRPV1 sensitizers. Amongst the strongest TRPV1 sensitizers were proteasome inhibitors, a class that includes bortezomib, a chemotherapeutic agent that causes small fiber neuropathy in 30-50% of patients. Prolonged exposure of bortezomib produced a TRPV1 sensitization that lasted several days and neurite retraction in vitro and histological and behavioral changes in male mice in vivo. TRPV1 knockout mice were protected from epidermal nerve fiber loss and a loss of sensory discrimination after bortezomib treatment. We conclude that long-term TRPV1 sensitization contributes to the development of bortezomib-induced neuropathy and the consequent loss of sensation, major deficits experienced by patients under this chemotherapeutic agent.
Collapse
Affiliation(s)
- Jared M Sprague
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Circle, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA, USA
| | - Ajay S Yekkirala
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Circle, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA, USA
| | - Bhagat Singh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Circle, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA, USA
| | - Ivan Tochitsky
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Circle, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA, USA
| | - Michael Stephens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Circle, Boston, MA, USA
| | - Octavio Viramontes
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Circle, Boston, MA, USA
| | - Jelena Ivanis
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Circle, Boston, MA, USA
| | - Natalia P Biscola
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leif A Havton
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Circle, Boston, MA, USA.
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA, USA.
| | - Alban Latremoliere
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Zafeiropoulou K, Kalampounias G, Alexis S, Androutsopoulou T, Katsoris P, Symeonidis A. Lower-Risk Myelodysplastic Syndrome (MDS) Patients Exhibit Diminished Proteasome Proteolytic Activity and High Intracellular Reactive Oxygen Species (ROS) Levels. Cureus 2023; 15:e49843. [PMID: 38169896 PMCID: PMC10758539 DOI: 10.7759/cureus.49843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2023] [Indexed: 01/05/2024] Open
Abstract
Myelodysplastic syndromes (MDS) constitute a heterogeneous group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis and an elevated risk of transformation to acute myeloid leukemia (AML). Available disease-modifying treatment approaches are limited. The ineffectiveness of proteasome inhibitors (PIs) in MDS patients is currently investigated, although it is unclear whether they rapidly develop resistance to PIs or whether proteasome proteolytic activity (PPA) is constitutively lower in the hematopoietic cells of these patients, thus limiting treatment effectiveness. We investigated 20 patients with MDS, categorized according to the International Prognostic Scoring System (IPSS) into a lower- or a higher-risk group. Peripheral blood mononuclear cells, bone marrow mononuclear cells, and cluster of differentiation 34-positive (CD34+) cells were isolated and assessed for the chymotrypsin-like activity of the proteasome and β5 subunit accumulation. Additionally, intracellular reactive oxygen species (ROS) generation was screened. The lower-risk patient group (n=10) exhibited significantly lower proteasome activity (p<0.001) compared to both the higher-risk group (n=10) and healthy subjects (n=10). Furthermore, the lower-risk group had elevated oxidative stress levels (p<0.0001) and reduced β5 subunit expression (p=0.0286). Both parameters were shown to be associated with transfusion dependency, since transfusion-dependent patients (n=5 in each subgroup) had decreased proteasome activity and simultaneously exhibited higher ROS levels. Our results indicate that reduced β5 expression might potentially explain PIs' ineffectiveness in lower-risk MDS, elucidating the importance of the risk group in the selection of the proper treatment algorithm.
Collapse
Affiliation(s)
| | | | | | | | | | - Argiris Symeonidis
- School of Medicine, University of Patras, Patras, GRC
- Hematology Division, Department of Internal Medicine, University General Hospital of Patras, Patras, GRC
| |
Collapse
|
4
|
Muto S, Matsubara T, Inoue T, Kitamura H, Yamamoto K, Ishii T, Yazawa M, Yamamoto R, Okada N, Mori K, Yamada H, Kuwabara T, Yonezawa A, Fujimaru T, Kawano H, Yokoi H, Doi K, Hoshino J, Yanagita M. Chapter 1: Evaluation of kidney function in patients undergoing anticancer drug therapy, from clinical practice guidelines for the management of kidney injury during anticancer drug therapy 2022. Int J Clin Oncol 2023; 28:1259-1297. [PMID: 37382749 DOI: 10.1007/s10147-023-02372-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/14/2023] [Indexed: 06/30/2023]
Abstract
The prevalence of CKD may be higher in patients with cancer than in those without due to the addition of cancer-specific risk factors to those already present for CKD. In this review, we describe the evaluation of kidney function in patients undergoing anticancer drug therapy. When anticancer drug therapy is administered, kidney function is evaluated to (1) set the dose of renally excretable drugs, (2) detect kidney disease associated with the cancer and its treatment, and (3) obtain baseline values for long-term monitoring. Owing to some requirements for use in clinical practice, a GFR estimation method such as the Cockcroft-Gault, MDRD, CKD-EPI, and the Japanese Society of Nephrology's GFR estimation formula has been developed that is simple, inexpensive, and provides rapid results. However, an important clinical question is whether they can be used as a method of GFR evaluation in patients with cancer. When designing a drug dosing regimen in consideration of kidney function, it is important to make a comprehensive judgment, recognizing that there are limitations regardless of which estimation formula is used or if GFR is directly measured. Although CTCAEs are commonly used as criteria for evaluating kidney disease-related adverse events that occur during anticancer drug therapy, a specialized approach using KDIGO criteria or other criteria is required when nephrologists intervene in treatment. Each drug is associated with the different disorders related to the kidney. And various risk factors for kidney disease associated with each anticancer drug therapy.
Collapse
Affiliation(s)
- Satoru Muto
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo, Japan.
| | - Takeshi Matsubara
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takamitsu Inoue
- Department of Renal and Urologic Surgery, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Hiroshi Kitamura
- Department of Urology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | | | - Taisuke Ishii
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Masahiko Yazawa
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | - Ryohei Yamamoto
- Department of Urology, Akita University Graduate School of Medicine, Akita, Japan
| | - Naoto Okada
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
- Pharmacy Department, Yamaguchi University Hospital, Yamaguchi, Japan
| | - Kiyoshi Mori
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Hiroyuki Yamada
- Department of Primary Care and Emergency Medicine, Kyoto University Hospital, Kyoto, Japan
| | - Takashige Kuwabara
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Atsushi Yonezawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Takuya Fujimaru
- Department of Nephrology, St Luke's International Hospital, Tokyo, Japan
| | - Haruna Kawano
- Department of Urology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hideki Yokoi
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kent Doi
- Department of Emergency and Critical Care Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Junichi Hoshino
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| |
Collapse
|
5
|
Byers HA, Brooks AN, Vangala JR, Grible JM, Feygin A, Clevenger CV, Harrell JC, Radhakrishnan SK. Evaluation of the NRF1-proteasome axis as a therapeutic target in breast cancer. Sci Rep 2023; 13:15843. [PMID: 37739987 PMCID: PMC10516926 DOI: 10.1038/s41598-023-43121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/20/2023] [Indexed: 09/24/2023] Open
Abstract
Proteasomes are multi-subunit complexes that specialize in protein degradation. Cancer cells exhibit a heightened dependence on proteasome activity, presumably to support their enhanced proliferation and other cancer-related characteristics. Here, a systematic analysis of TCGA breast cancer datasets revealed that proteasome subunit transcript levels are elevated in all intrinsic subtypes (luminal, HER2-enriched, and basal-like/triple-negative) when compared to normal breast tissue. Although these observations suggest a pan-breast cancer utility for proteasome inhibitors, our further experiments with breast cancer cell lines and patient-derived xenografts (PDX) pointed to triple-negative breast cancer (TNBC) as the most sensitive subtype to proteasome inhibition. Finally, using TNBC cells, we extended our studies to in vivo xenograft experiments. Our previous work has firmly established a cytoprotective role for the transcription factor NRF1 via its ability to upregulate proteasome genes in response to proteasome inhibition. In further support of this notion, we show here that NRF1 depletion significantly reduced tumor burden in an MDA-MB-231 TNBC xenograft mouse model treated with carfilzomib. Taken together, our results point to TNBC as a particularly vulnerable breast cancer subtype to proteasome inhibition and provide a proof-of-principle for targeting NRF1 as a viable means to increase the efficacy of proteasome inhibitors in TNBC tumors.
Collapse
Affiliation(s)
- Holly A Byers
- Department of Pathology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Amy N Brooks
- Department of Pathology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Janakiram R Vangala
- Department of Pathology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Jacqueline M Grible
- Department of Pathology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Alex Feygin
- Department of Pathology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Charles V Clevenger
- Department of Pathology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - J Chuck Harrell
- Department of Pathology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Senthil K Radhakrishnan
- Department of Pathology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
6
|
Sanati M, Afshari AR, Ahmadi SS, Moallem SA, Sahebkar A. Modulation of the ubiquitin-proteasome system by phytochemicals: Therapeutic implications in malignancies with an emphasis on brain tumors. Biofactors 2023; 49:782-819. [PMID: 37162294 DOI: 10.1002/biof.1958] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/20/2023] [Indexed: 05/11/2023]
Abstract
Regarding the multimechanistic nature of cancers, current chemo- or radiotherapies often fail to eradicate disease pathology, and frequent relapses or resistance to therapies occur. Brain malignancies, particularly glioblastomas, are difficult-to-treat cancers due to their highly malignant and multidimensional biology. Unfortunately, patients suffering from malignant tumors often experience poor prognoses and short survival periods. Thus far, significant efforts have been conducted to discover novel and more effective modalities. To that end, modulation of the ubiquitin-proteasome system (UPS) has attracted tremendous interest since it affects the homeostasis of proteins critically engaged in various cell functions, for example, cell metabolism, survival, proliferation, and differentiation. With their safe and multimodal actions, phytochemicals are among the promising therapeutic tools capable of turning the operation of various UPS elements. The present review, along with an updated outline of the role of UPS dysregulation in multiple cancers, provided a detailed discussion on the impact of phytochemicals on the UPS function in malignancies, especially brain tumors.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
- Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Amir R Afshari
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam-Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Adel Moallem
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Zahraa University for Women, Karbala, Iraq
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Matsumoto T, Matsumoto J, Matsushita Y, Arimura M, Aono K, Aoki M, Terada K, Mori M, Haramaki Y, Imatoh T, Yamauchi A, Migita K. Bortezomib Increased Vascular Permeability by Decreasing Cell-Cell Junction Molecules in Human Pulmonary Microvascular Endothelial Cells. Int J Mol Sci 2023; 24:10842. [PMID: 37446020 DOI: 10.3390/ijms241310842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Bortezomib (BTZ), a chemotherapeutic drug used to treat multiple myeloma, induces life-threatening side effects, including severe pulmonary toxicity. However, the mechanisms underlying these effects remain unclear. The objectives of this study were to (1) investigate whether BTZ influences vascular permeability and (2) clarify the effect of BTZ on the expression of molecules associated with cell-cell junctions using human pulmonary microvascular endothelial cells in vitro. Clinically relevant concentrations of BTZ induced limited cytotoxicity and increased the permeability of human pulmonary microvascular endothelial cell monolayers. BTZ decreased the protein expression of claudin-5, occludin, and VE-cadherin but not that of ZO-1 and β-catenin. Additionally, BTZ decreased the mRNA expression of claudin-5, occludin, ZO-1, VE-cadherin, and β-catenin. Our results suggest that BTZ increases the vascular permeability of the pulmonary microvascular endothelium by downregulating cell-cell junction molecules, particularly claudin-5, occludin, and VE-cadherin.
Collapse
Affiliation(s)
- Taichi Matsumoto
- Basic Medical Research Unit, St. Mary's Research Center, 422, Tsubuku-honmachi, Kurume 830-8543, Fukuoka, Japan
| | - Junichi Matsumoto
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Fukuoka, Japan
| | - Yuka Matsushita
- Department of Drug Informatics and Translational Research, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Fukuoka, Japan
| | - Moeno Arimura
- Department of Drug Informatics and Translational Research, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Fukuoka, Japan
| | - Kentaro Aono
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Fukuoka, Japan
| | - Mikiko Aoki
- Department of Pathology, Faculty of Medicine, Fukuoka University, 7-45-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Fukuoka, Japan
| | - Kazuki Terada
- Division of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 7-2-1, Kamiohno, Himeji 670-8524, Hyogo, Japan
| | - Masayoshi Mori
- Department of Pharmacotherapeutics, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Fukuoka, Japan
| | - Yutaka Haramaki
- Psychology Program, Graduate School of Humanities and Social Sciences, Hiroshima University, Kagamiyama, 1-1-1, Kagamiyama, Higashi-Hiroshima City 739-8512, Hiroshima, Japan
| | - Takuya Imatoh
- Department of Drug Informatics and Translational Research, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Fukuoka, Japan
| | - Atsushi Yamauchi
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Fukuoka, Japan
| | - Keisuke Migita
- Department of Drug Informatics and Translational Research, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1, Nanakuma, Jonan-ku, Fukuoka 814-0180, Fukuoka, Japan
| |
Collapse
|
8
|
Munir A, Fazal S, Bhatti AI, Sajjad B. Pharmacokinetics-Pharmacodynamics Modeling and Evaluation of Tumor Response to Bortezomib Proteasome Inhibitor in Waldenstrom Macroglobulinemia. Am J Clin Oncol 2023; 46:150-160. [PMID: 36808095 DOI: 10.1097/coc.0000000000000982] [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: 02/22/2023]
Abstract
BACKGROUND Waldenstrom's macroglobulinemia (WM), also known as lymphoplasmacytic lymphoma, is a type of non-Hodgkin's lymphoma in which the malignant cells produce many macroglobulin proteins. It originates from B cells and develops in the bone marrow, where Wm cells combine to produce distinct types of blood cells, resulting in reduced volumes of red blood cells, white blood cells, and platelets, making it harder for the body to fight diseases. Chemoimmunotherapy is being used for the clinical management of WM, but new targeted agents, the BTK inhibitor ibrutinib and the proteasome inhibitor bortezomib, have shown significant improvements in patients with relapsed/refractory WM. However, given its effectiveness, drug resistance and relapse are normal, and there is little research on the pathways responsible for drug effects on the tumor. METHODS In this study, Pharmacokinetics-pharmacodynamic simulations were done to assess the effect of the proteasome inhibitor bortezomib on the tumor. For this purpose, the Pharmacokinetics-pharmacodynamic model was developed. The model parameters were determined and calculated using the Ordinary Differential Equation solver toolbox and the least-squares function. Pharmacokinetic profiles and pharmacodynamic analysis were performed to determine the change in tumor weight associated with the use of proteasome inhibitors. RESULTS Bortezomib and ixazomib have been found to reduce tumor weight briefly, but once the dose is reduced, the tumor begins to grow again. Carfilzomib and oprozomib had better results, and rituximab reduced tumor weight more effectively. CONCLUSION Once validated, it is proposed that a combination of selected drugs can be evaluated in the laboratory to treat WM.
Collapse
Affiliation(s)
- Anum Munir
- School of Biosciences, University of Kent, Canterbury
- Department of Applied Biology, The London College UCK, London
| | - Sahar Fazal
- Department of Bioinformatics and Biosciences, Faculty of Health, and Life Sciences
| | - Aamer Iqbal Bhatti
- Department of Electrical Engineering, Capital University of Science and Technology, Islamabad
| | - Bilawal Sajjad
- Smart Sec Solutions, London
- Department of Information Technology, Hazara University Mansehra, Pakistan
| |
Collapse
|
9
|
Matsubara T, Yokoi H, Yamada H, Yanagita M. Nephrotoxicity associated with anticancer agents: perspective on onconephrology from nephrologists. Int J Clin Oncol 2023; 28:625-636. [PMID: 36872414 DOI: 10.1007/s10147-023-02307-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/01/2023] [Indexed: 03/07/2023]
Abstract
Nephrotoxicity is one of the most important complications in cancer patients. In particular, acute kidney injury (AKI) is known to be associated with discontinuing effective oncological treatments, longer hospitalizations, increased costs, and a higher risk of death. In addition to acute kidney injury, clinical signs associated with nephrotoxicity during treatment with anticancer agents include chronic kidney disease, proteinuria, hypertension, electrolyte abnormalities, and other characteristic manifestations. Many of these signs are caused both by cancer treatment as well as by cancer itself. Therefore, it is important to carefully recognize whether the underlying causes of renal impairment in cancer patients are cancer-related, treatment-related, or both. This review describes the epidemiology and pathophysiology of anticancer agent-induced acute kidney injury, proteinuria, hypertension, and other characteristic manifestations.
Collapse
Affiliation(s)
- Takeshi Matsubara
- Department of Nephrology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Hideki Yokoi
- Department of Nephrology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroyuki Yamada
- Department of Primary Care and Emergency Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| |
Collapse
|
10
|
Intermediate-dose cyclophosphamide and bortezomib for PBSC mobilization in multiple myeloma. Transfus Apher Sci 2023:103649. [PMID: 36739175 DOI: 10.1016/j.transci.2023.103649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Although the incorporation of bortezomib into induction regimens has improved, response rates in patients with multiple myeloma (MM), the role of bortezomib in the, peripheral blood stem cell (PBSC) mobilization remains unclear. We assessed the, PBSC mobilization efficacy, safety, and disease response of intermediate-dose, cyclophosphamide and bortezomib in the PBSC mobilization. Twenty-one patients with, newly diagnosed MM were enrolled in a phase II, non-randomized study that used, bortezomib (1.3 mg/m2/day on days 1, 4, 8, and 11) and intermediate-dose, cyclophosphamide (2 g/m2/day on days 2, 3) (Bor-ID-CY). The data from 15 patients, who received intermediate-dose cyclophosphamide (ID-CY) were used as a historical, control group. The total CD34 + cell yield of Bor-ID-CY and ID-CY groups were not, significantly different (median 6.3 ×106/kg vs. 6.5 ×106/kg, p = 0.19). All three patients, with mobilization failure of two groups had t(11;14). Six patients in Bor-ID-CY group, were upgraded from a status that was less than a very good partial response (VGPR), at the time of PBSC mobilization to a VGPR or better after PBSC mobilization, (p = 0.014). Four patients in Bor-ID-CY group developed sepsis. The time to, engraftment was similar in the two groups. The addition of bortezomib to ID-CY did not, impact the stem cell yield or quality.
Collapse
|
11
|
Rani S, Sahoo RK, Kumar V, Chaurasiya A, Kulkarni O, Mahale A, Katke S, Kuche K, Yadav V, Jain S, Nakhate KT, Ajazuddin, Gupta U. N-2-Hydroxypropylmethacrylamide-Polycaprolactone Polymeric Micelles in Co-delivery of Proteasome Inhibitor and Polyphenol: Exploration of Synergism or Antagonism. Mol Pharm 2023; 20:524-544. [PMID: 36306447 DOI: 10.1021/acs.molpharmaceut.2c00752] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Breast cancer leads to the highest mortality among women resulting in a major clinical burden. Multidrug therapy is more efficient in such patients compared to monodrug therapy. Simultaneous combinatorial or co-delivery garnered significant interest in the past years. Caffeic acid (CFA) (a natural polyphenol) has received growing attention because of its anticarcinogenic and antioxidant potential. Bortezomib (BTZ) is a proteasome inhibitor and may be explored for treating breast cancer. Despite its high anticancer activity, the low water solubility and chemical instability restrict its efficacy against solid tumors. In the present study, we designed and investigated a HP-PCL (N-2-hydroxypropylmethacrylamide-polycaprolactone) polymeric micellar (PMCs) system for the simultaneous delivery of BTZ and CFA in the treatment of breast cancer. The designed BTZ+CFA-HP-PCL PMCs were fabricated, optimized, and characterized for size, zeta potential, surface morphology, and in vitro drug release. Developed nanosized (174.6 ± 0.24 nm) PMCs showed enhanced cellular internalization and cell cytotoxicity in both MCF-7 and MDA-MB-231 cells. ROS (reactive oxygen species) levels were highest in BTZ-HP-PCL PMCs, while CFA-HP-PCL PMCs significantly (p < 0.001) scavenged the ROS generated in 2',7'-dichlorofluorescein diacetate (DCFH-DA) assay. The mitochondrial membrane potential (MMP) assay revealed intense and significant green fluorescence in both types of cancer cells when treated with BTZ-HP-PCL PMCs (p < 0.001) indicating apoptosis or cell death. The pharmacokinetic studies revealed that BTZ-HP-PCL PMCs and BTZ+CFA-HP-PCL PMCs exhibited the highest bioavailability, enhanced plasma half-life, decreased volume of distribution, and lower clearance rate than the pure combination of drugs. In the organ biodistribution studies, the combination of BTZ+CFA showed higher distribution in the spleen and the heart. Overall findings of in vitro studies surprisingly resulted in better therapeutic efficiency of BTZ-HP-PCL PMCs than BTZ+CFA-HP-PCL PMCs. However, the in vivo tumor growth inhibition study performed in tumor-induced mice concluded that the tumor growth was inhibited by both BTZ-HP-PCL PMCs and BTZ+CFA-HP-PCL PMCs (p < 0.0001) more efficiently than pure BTZ and the combination (BTZ+CFA), which may be due to the conversion of boronate ester into boronic acid. Henceforth, the combination of BTZ and CFA provides further indications to be explored in the future to support the hypothesis that BTZ may work with polyphenol (CFA) in the acidic environment of the tumor.
Collapse
Affiliation(s)
- Sarita Rani
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan305817, India
| | - Rakesh K Sahoo
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan305817, India
| | - Vinay Kumar
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan305817, India
| | - Akash Chaurasiya
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal Medchal District, Hyderabad, Telangana500078, India
| | - Onkar Kulkarni
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal Medchal District, Hyderabad, Telangana500078, India
| | - Ashutosh Mahale
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal Medchal District, Hyderabad, Telangana500078, India
| | - Sumeet Katke
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal Medchal District, Hyderabad, Telangana500078, India
| | - Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, Punjab160062, India
| | - Vivek Yadav
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, Punjab160062, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar, Punjab160062, India
| | - Kartik T Nakhate
- Department of Pharmacology, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra424001, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh490024, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan305817, India
| |
Collapse
|
12
|
Targeting immunoproteasome in neurodegeneration: A glance to the future. Pharmacol Ther 2023; 241:108329. [PMID: 36526014 DOI: 10.1016/j.pharmthera.2022.108329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
The immunoproteasome is a specialized form of proteasome equipped with modified catalytic subunits that was initially discovered to play a pivotal role in MHC class I antigen processing and immune system modulation. However, over the last years, this proteolytic complex has been uncovered to serve additional functions unrelated to antigen presentation. Accordingly, it has been proposed that immunoproteasome synergizes with canonical proteasome in different cell types of the nervous system, regulating neurotransmission, metabolic pathways and adaptation of the cells to redox or inflammatory insults. Hence, studying the alterations of immunoproteasome expression and activity is gaining research interest to define the dynamics of neuroinflammation as well as the early and late molecular events that are likely involved in the pathogenesis of a variety of neurological disorders. Furthermore, these novel functions foster the perspective of immunoproteasome as a potential therapeutic target for neurodegeneration. In this review, we provide a brain and retina-wide overview, trying to correlate present knowledge on structure-function relationships of immunoproteasome with the variety of observed neuro-modulatory functions.
Collapse
|
13
|
Koerner J, Horvath D, Oliveri F, Li J, Basler M. Suppression of prostate cancer and amelioration of the immunosuppressive tumor microenvironment through selective immunoproteasome inhibition. Oncoimmunology 2022; 12:2156091. [PMID: 36531689 PMCID: PMC9757486 DOI: 10.1080/2162402x.2022.2156091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
New treatment options to battle hormone-refractory prostate carcinoma (PC) are a pressing medical need. Chronic inflammation has been implicated in PC etiology. The pro-inflammatory cytokines IL-6, IL-23 and IL-17 are key mediators to promote growth of PC. Here, we evaluate the potential of immunoproteasome inhibition for anti-inflammatory and direct anti-tumorigenic therapy of PC. The anti-tumor effect of immunoproteasome inhibitor ONX 0914 was tested in mouse and human PC cells and the in vivo therapeutic efficacy of immunoproteasome inhibition was analyzed in transgenic adenocarcinoma of the mouse prostate (TRAMP) mice in preventive and therapeutic settings and in castration-resistant (CR)PC after castration. Inhibition of the immunoproteasome subunit LMP7 induced apoptotic cell death in PC cell lines. In TRAMP mice, ONX 0914-treatment resulted in significant inhibition of PC growth with a decreased frequency of malignant prostatic lesions and inhibition of metastasis formation. The number of immunosuppressive myeloid cells in PC was greatly reduced in response to ONX 0914. Thus, immunoproteasome inhibition shows remarkable efficacy against PC progression in vivo and impedes tumor recurrence in CRPC-TRAMP mice by blocking the immunosuppressive inflammatory response in the tumor microenvironment. In conclusion, we show that the immunoproteasome is a promising drug target for the treatment of PC.
Collapse
Affiliation(s)
- Julia Koerner
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Dennis Horvath
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany,Centre for the Advanced Study of Collective Behaviour, University of Konstanz, Konstanz, Germany
| | - Franziska Oliveri
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Jun Li
- Department of Urologic Oncology Surgery, Chongqing University Cancer Hospital, Chongqing, China,Jun Li Department of Urologic Oncology Surgery, Chongqing University Cancer Hospital, Han Yu Road 181, 400030 Chongqing, China
| | - Michael Basler
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany,Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland,CONTACT Michael Basler Division of Immunology, Department of Biology, University of Konstanz, Universitaetsstr. 10, D-78457, Konstanz, Germany
| |
Collapse
|
14
|
Winter JM, Fresenius HL, Cunningham CN, Wei P, Keys HR, Berg J, Bott A, Yadav T, Ryan J, Sirohi D, Tripp SR, Barta P, Agarwal N, Letai A, Sabatini DM, Wohlever ML, Rutter J. Collateral deletion of the mitochondrial AAA+ ATPase ATAD1 sensitizes cancer cells to proteasome dysfunction. eLife 2022; 11:82860. [PMID: 36409067 DOI: 10.7554/elife.82860] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
Abstract
The tumor suppressor gene PTEN is the second most commonly deleted gene in cancer. Such deletions often include portions of the chromosome 10q23 locus beyond the bounds of PTEN itself, which frequently disrupts adjacent genes. Coincidental loss of PTEN-adjacent genes might impose vulnerabilities that could either affect patient outcome basally or be exploited therapeutically. Here, we describe how the loss of ATAD1, which is adjacent to and frequently co-deleted with PTEN, predisposes cancer cells to apoptosis triggered by proteasome dysfunction and correlates with improved survival in cancer patients. ATAD1 directly and specifically extracts the pro-apoptotic protein BIM from mitochondria to inactivate it. Cultured cells and mouse xenografts lacking ATAD1 are hypersensitive to clinically used proteasome inhibitors, which activate BIM and trigger apoptosis. This work furthers our understanding of mitochondrial protein homeostasis and could lead to new therapeutic options for the hundreds of thousands of cancer patients who have tumors with chromosome 10q23 deletion.
Collapse
Affiliation(s)
- Jacob M Winter
- Department of Biochemistry, University of Utah, Salt Lake City, United States
| | - Heidi L Fresenius
- Department of Chemistry & Biochemistry, University of Toledo, Toledo, United States
| | - Corey N Cunningham
- Department of Biochemistry, University of Utah, Salt Lake City, United States
| | - Peng Wei
- Department of Biochemistry, University of Utah, Salt Lake City, United States
| | - Heather R Keys
- Whitehead Institute for Biomedical Research, Cambridge, United States
| | - Jordan Berg
- Department of Biochemistry, University of Utah, Salt Lake City, United States
| | - Alex Bott
- Department of Biochemistry, University of Utah, Salt Lake City, United States
| | - Tarun Yadav
- Department of Biochemistry, University of Utah, Salt Lake City, United States
| | - Jeremy Ryan
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States
| | - Deepika Sirohi
- University of Utah and ARUP Laboratories, Salt Lake City, United States
| | - Sheryl R Tripp
- University of Utah and ARUP Laboratories, Salt Lake City, United States
| | - Paige Barta
- Department of Biochemistry, University of Utah, Salt Lake City, United States
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, United States
| | - Anthony Letai
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, United States
| | - David M Sabatini
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Matthew L Wohlever
- Department of Chemistry & Biochemistry, University of Toledo, Toledo, United States
| | - Jared Rutter
- Department of Biochemistry, University of Utah, Salt Lake City, United States.,Huntsman Cancer Institute, University of Utah, Salt Lake City, United States.,Howard Hughes Medical Institute, Salt Lake City, United States
| |
Collapse
|
15
|
Holzer AK, Suciu I, Karreman C, Goj T, Leist M. Specific Attenuation of Purinergic Signaling during Bortezomib-Induced Peripheral Neuropathy In Vitro. Int J Mol Sci 2022; 23:ijms23073734. [PMID: 35409095 PMCID: PMC8998302 DOI: 10.3390/ijms23073734] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 12/18/2022] Open
Abstract
Human peripheral neuropathies are poorly understood, and the availability of experimental models limits further research. The PeriTox test uses immature dorsal root ganglia (DRG)-like neurons, derived from induced pluripotent stem cells (iPSC), to assess cell death and neurite damage. Here, we explored the suitability of matured peripheral neuron cultures for the detection of sub-cytotoxic endpoints, such as altered responses of pain-related P2X receptors. A two-step differentiation protocol, involving the transient expression of ectopic neurogenin-1 (NGN1) allowed for the generation of homogeneous cultures of sensory neurons. After >38 days of differentiation, they showed a robust response (Ca2+-signaling) to the P2X3 ligand α,β-methylene ATP. The clinical proteasome inhibitor bortezomib abolished the P2X3 signal at ≥5 nM, while 50−200 nM was required in the PeriTox test to identify neurite damage and cell death. A 24 h treatment with low nM concentrations of bortezomib led to moderate increases in resting cell intracellular Ca2+ concentration but signaling through transient receptor potential V1 (TRPV1) receptors or depolarization-triggered Ca2+ influx remained unaffected. We interpreted the specific attenuation of purinergic signaling as a functional cell stress response. A reorganization of tubulin to form dense structures around the cell somata confirmed a mild, non-cytotoxic stress triggered by low concentrations of bortezomib. The proteasome inhibitors carfilzomib, delanzomib, epoxomicin, and MG-132 showed similar stress responses. Thus, the model presented here may be used for the profiling of new proteasome inhibitors in regard to their side effect (neuropathy) potential, or for pharmacological studies on the attenuation of their neurotoxicity. P2X3 signaling proved useful as endpoint to assess potential neurotoxicants in peripheral neurons.
Collapse
Affiliation(s)
- Anna-Katharina Holzer
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany; (A.-K.H.); (I.S.); (C.K.); (T.G.)
| | - Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany; (A.-K.H.); (I.S.); (C.K.); (T.G.)
- Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, 78457 Konstanz, Germany
| | - Christiaan Karreman
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany; (A.-K.H.); (I.S.); (C.K.); (T.G.)
| | - Thomas Goj
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany; (A.-K.H.); (I.S.); (C.K.); (T.G.)
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, 78457 Konstanz, Germany; (A.-K.H.); (I.S.); (C.K.); (T.G.)
- CAAT-Europe, University of Konstanz, 78457 Konstanz, Germany
- Correspondence: ; Tel.: +49-(0)-7531-88-5037
| |
Collapse
|
16
|
Goel L, Das US, Gupta P, Velpandian T, Kumar L, Singh A, Luthra K, Gupta YK. Development and Validation of Highly Sensitive LC–ESI-MS/MS Method for Bortezomib and Its Applications for Plasma Levels and Drug Content of Branded and Generic Formulations in India. Chromatographia 2022. [DOI: 10.1007/s10337-021-04121-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
17
|
Liu J, Zhao R, Jiang X, Li Z, Zhang B. Progress on the Application of Bortezomib and Bortezomib-Based Nanoformulations. Biomolecules 2021; 12:biom12010051. [PMID: 35053199 PMCID: PMC8773474 DOI: 10.3390/biom12010051] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022] Open
Abstract
Bortezomib (BTZ) is the first proteasome inhibitor approved by the Food and Drug Administration. It can bind to the amino acid residues of the 26S proteasome, thereby causing the death of tumor cells. BTZ plays an irreplaceable role in the treatment of mantle cell lymphoma and multiple myeloma. Moreover, its use in the treatment of other hematological cancers and solid tumors has been investigated in numerous clinical trials and preclinical studies. Nevertheless, the applications of BTZ are limited due to its insufficient specificity, poor permeability, and low bioavailability. Therefore, in recent years, different BTZ-based drug delivery systems have been evaluated. In this review, we firstly discussed the functions of proteasome inhibitors and their mechanisms of action. Secondly, the properties of BTZ, as well as recent advances in both clinical and preclinical research, were reviewed. Finally, progress in research regarding BTZ-based nanoformulations was summarized.
Collapse
Affiliation(s)
| | | | | | | | - Bo Zhang
- Correspondence: ; Tel.: +86-636-8462490
| |
Collapse
|
18
|
Wang J, Fang Y, Fan RA, Kirk CJ. Proteasome Inhibitors and Their Pharmacokinetics, Pharmacodynamics, and Metabolism. Int J Mol Sci 2021; 22:ijms222111595. [PMID: 34769030 PMCID: PMC8583966 DOI: 10.3390/ijms222111595] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
The proteasome is responsible for mediating intracellular protein degradation and regulating cellular function with impact on tumor and immune effector cell biology. The proteasome is found predominantly in two forms, the constitutive proteasome and the immunoproteasome. It has been validated as a therapeutic drug target through regulatory approval with 2 distinct chemical classes of small molecular inhibitors (boronic acid derivatives and peptide epoxyketones), including 3 compounds, bortezomib (VELCADE), carfilzomib (KYPROLIS), and ixazomib (NINLARO), for use in the treatment of the plasma cell neoplasm, multiple myeloma. Additionally, a selective inhibitor of immunoproteasome (KZR-616) is being developed for the treatment of autoimmune diseases. Here, we compare and contrast the pharmacokinetics (PK), pharmacodynamics (PD), and metabolism of these 2 classes of compounds in preclinical models and clinical studies. The distinct metabolism of peptide epoxyketones, which is primarily mediated by microsomal epoxide hydrolase, is highlighted and postulated as a favorable property for the development of this class of compound in chronic conditions.
Collapse
|
19
|
Guo ZX, Yang F, van den Anker JN, Zheng Y, Zhao W. A simplified method for bortezomib determination using dried blood spots in combination with liquid chromatography/tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1181:122905. [PMID: 34438277 DOI: 10.1016/j.jchromb.2021.122905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/12/2021] [Accepted: 08/20/2021] [Indexed: 11/30/2022]
Abstract
Bortezomib, a proteinase inhibitor currently used to treat multiple myeloma and mantle cell lymphoma, has a high incidence of adverse reactions and large inter-individual differences in plasma concentrations. A simple, validated LC-MS/MS method for the quantitative analysis of bortezomib in dried blood spot (DBS) samples was developed to provide support for determining the effective concentration range of bortezomib for clinical use. Fifty (i50) μL of spiked blood were added onto Whatman protein saver cards to prepare the DBS samples. Circular cards of 6 mm diameter were punched, extracted by methanol containing the internal standard (apatinib), and injected into the LC-MS/MS system. The method validation included selectivity, linearity, accuracy and precision, stability, matrix effect, recovery and hematocrit. The calibration curve showed correlation coefficient values higher than 0.999 in the range of 0.2 - 20.0 ng/mL for bortezomib. The acceptance criteria of accuracy (relative error < 12.5%) and precision (coefficient of variation < 10.7%) were met in all cases. The matrix effect was<13.2%, and the recovery was between 87.3 and 100.2%. DBS samples were shown to be stable when stored in cold conditions or at room temperature. Different hematocrit values did not significantly affect the accuracy of the measured concentrations. And there are no significant differences between bortezomib concentrations in DBS samples and plasma samples. This new method was successfully used for clinical concentration determinations of bortezomib and can be applied in future therapeutic drug monitoring and pharmacokinetic studies of bortezomib especially in pediatric patients.
Collapse
Affiliation(s)
- Zi-Xuan Guo
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fan Yang
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Johannes N van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, WA, DC, USA; Departments of Pediatrics, Pharmacology & Physiology, Genomics & Precision Medicine, George Washington University, School of Medicine and Health Sciences, WA, DC, USA; Department of Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, University of Basel, Switzerland
| | - Yi Zheng
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Wei Zhao
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China; Department of Clinical Pharmacy, Clinical Trial Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan, China.
| |
Collapse
|
20
|
Molecular analysis of cell survival and death pathways in the proteasome inhibitor bortezomib-resistant PC3 prostate cancer cell line. Med Oncol 2021; 38:112. [PMID: 34363546 DOI: 10.1007/s12032-021-01563-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/02/2021] [Indexed: 11/09/2022]
Abstract
The ubiquitin-proteasome pathway is an important protein quality control system involved in intracellular homeostasis. To achieve intracellular homeostasis, proteins that are misfolded as a result of translational errors or genetic mutations must be eliminated by the ubiquitin-proteasome pathway. In our previous publications, we determined that 4T1 breast and B16F10 melanoma cancer cells have differential levels of resistance to proteasome inhibitors. Again, in the previous studies, we reported that 4T1 cell cultures, despite being p53-mutant, underwent apoptosis as a result of bortezomib treatment. The first goal of this study was to verify the resistance levels of parental and resistant PC3 prostate cancer cells to bortezomib using WST-1 test. As a result of treatment with different bortezomib concentrations for 48 h, the IC50 value of the parental cells was determined as 32.8 nM and that of the resistant cells was determined as 346 nM. This result showed that the resistant cells were at least 10.5 times more resistant. In addition, to determine whether the resistance gained was reversible or not, the cells were passaged in a medium without bortezomib for one month. The IC50 value determination by WST-1 test showed that the resistant PC3 cells gained an irreversible bortezomib resistance phenotype. The results of the 3D spheroid experiment showed that the 3D spheroid diameter of resistant cells was significantly higher than that of the parental cells. The studies conducted with Western blot showed that ERK1 MAPK T202 phosphorylation and the conversion of autophagy marker LC3-I to LC3-II were significantly increased in parental cells as compared to the resistant cells. Finally, the results showed that while both maternal embryonic leucine zipper kinase (MELK) inhibitor OTSSP167 and Ca2+ chelator BAPTA-AM (also an inhibitor of the expression of antiapoptotic protein GRP78) are promising agents for cancer cells resistant to the proteasome inhibitors, CDK2 inhibitor CVT-313 was found ineffective in both parental and the resistant cells.
Collapse
|
21
|
Penke LRK, Speth J, Wettlaufer S, Draijer C, Peters-Golden M. Bortezomib Inhibits Lung Fibrosis and Fibroblast Activation Without Proteasome Inhibition. Am J Respir Cell Mol Biol 2021; 66:23-37. [PMID: 34236953 DOI: 10.1165/rcmb.2021-0112oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The FDA-approved proteasomal inhibitor bortezomib (BTZ) has attracted interest for its potential anti-fibrotic actions. However, neither its in vivo efficacy in lung fibrosis nor its dependence on proteasome inhibition has been conclusively defined. In this study, we assessed the therapeutic efficacy of BTZ in a mouse model of pulmonary fibrosis, developed an in vitro protocol to define its actions on diverse fibroblast activation parameters, determined its reliance on proteasome inhibition for these actions in vivo and in vitro and explored alternative mechanisms of action. The therapeutic administration of BTZ diminished the severity of pulmonary fibrosis without reducing proteasome activity in the lung. In experiments designed to mimic this lack of proteasome inhibition in vitro, BTZ reduced fibroblast proliferation, differentiation into myofibroblasts, and collagen synthesis. It promoted de-differentiation of myofibroblasts and overcame their characteristic resistance to apoptosis. Mechanistically, BTZ inhibited kinases important for fibroblast activation while inducing expression of dual-specificity phosphatase 1 or DUSP1, and knockdown of DUSP1 abolished its anti-fibrotic actions in fibroblasts. Collectively, these findings suggest that BTZ exhibits a multidimensional profile of robust inhibitory actions on lung fibroblasts as well as anti-fibrotic actions in vivo. Unexpectedly, these actions appear to be independent of proteasome inhibition, and instead attributable to induction of DUSP1.
Collapse
Affiliation(s)
| | - Jennifer Speth
- University of Michigan, 1259, Ann Arbor, Michigan, United States
| | - Scott Wettlaufer
- University of Michigan, 1259, Division of Pulmonary and Critical Care Medicine, Ann Arbor, Michigan, United States
| | | | - Marc Peters-Golden
- University of Michigan Health System, 21707, Ann Arbor, Michigan, United States;
| |
Collapse
|
22
|
Northrop A, Byers HA, Radhakrishnan SK. Regulation of NRF1, a master transcription factor of proteasome genes: implications for cancer and neurodegeneration. Mol Biol Cell 2021; 31:2158-2163. [PMID: 32924844 PMCID: PMC7550695 DOI: 10.1091/mbc.e20-04-0238] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The ability to sense proteasome insufficiency and respond by directing the transcriptional synthesis of de novo proteasomes is a trait that is conserved in evolution and is found in organisms ranging from yeast to humans. This homeostatic mechanism in mammalian cells is driven by the transcription factor NRF1. Interestingly, NRF1 is synthesized as an endoplasmic reticulum (ER) membrane protein and when cellular proteasome activity is sufficient, it is retrotranslocated into the cytosol and targeted for destruction by the ER-associated degradation pathway (ERAD). However, when proteasome capacity is diminished, retrotranslocated NRF1 escapes ERAD and is activated into a mature transcription factor that traverses to the nucleus to induce proteasome genes. In this Perspective, we track the journey of NRF1 from the ER to the nucleus, with a special focus on the various molecular regulators it encounters along its way. Also, using human pathologies such as cancer and neurodegenerative diseases as examples, we explore the notion that modulating the NRF1-proteasome axis could provide the basis for a viable therapeutic strategy in these cases.
Collapse
Affiliation(s)
- Amy Northrop
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298
| | - Holly A Byers
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298
| | | |
Collapse
|
23
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
24
|
Tan CRC, Abdul-Majeed S, Cael B, Barta SK. Clinical Pharmacokinetics and Pharmacodynamics of Bortezomib. Clin Pharmacokinet 2020; 58:157-168. [PMID: 29802543 DOI: 10.1007/s40262-018-0679-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteasome inhibitors disrupt multiple pathways in cells and the bone marrow microenvironment, resulting in apoptosis and inhibition of cell-cycle progression, angiogenesis, and proliferation. Bortezomib is a first-in-class proteasome inhibitor approved for the treatment of multiple myeloma and mantle cell lymphoma after one prior therapy. It is also effective in other plasma cell disorders and non-Hodgkin lymphomas. The main mechanism of action of bortezomib is to inhibit the chymotrypsin-like site of the 20S proteolytic core within the 26S proteasome, thereby inducing cell-cycle arrest and apoptosis. The pharmacokinetic profile of intravenous bortezomib is characterized by a two-compartment model with a rapid initial distribution phase followed by a longer elimination phase and a large volume of distribution. Bortezomib is available for subcutaneous and intravenous administration. Pharmacokinetic studies comparing subcutaneous and intravenous bortezomib demonstrated that systemic exposure was equivalent for both routes; pharmacodynamic parameters of 20S proteasome inhibition were also similar. Renal impairment does not influence the intrinsic pharmacokinetics of bortezomib. However, moderate or severe hepatic impairment causes an increase in plasma concentrations of bortezomib. Therefore, patients with moderate or severe hepatic impairment should start at a reduced dose. Because bortezomib undergoes extensive metabolism by hepatic cytochrome P450 3A4 and 2C19 enzymes, certain strong cytochrome P450 3A4 inducers and inhibitors can also alter the systemic exposure of bortezomib. This article critically reviews and summarizes the clinical pharmacokinetics and pharmacodynamics of bortezomib at various dosing levels and routes of administration as well as in specific patient subsets. In addition, we discuss the clinical efficacy and safety of bortezomib.
Collapse
Affiliation(s)
- Carlyn Rose C Tan
- Department of Hematology/Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Saif Abdul-Majeed
- Office of Clinical Research, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Brittany Cael
- Department of Pharmacy, Bone Marrow Transplant Program, Jeanes Hospital, Philadelphia, PA, USA
| | - Stefan K Barta
- Department of Hematology/Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA.
| |
Collapse
|
25
|
Raninga PV, Lee A, Sinha D, Dong LF, Datta KK, Lu X, Kalita-de Croft P, Dutt M, Hill M, Pouliot N, Gowda H, Kalimutho M, Neuzil J, Khanna KK. Marizomib suppresses triple-negative breast cancer via proteasome and oxidative phosphorylation inhibition. Am J Cancer Res 2020; 10:5259-5275. [PMID: 32373211 PMCID: PMC7196287 DOI: 10.7150/thno.42705] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/11/2020] [Indexed: 01/05/2023] Open
Abstract
Purpose: Lacking effective targeted therapies, triple-negative breast cancer (TNBCs) is highly aggressive and metastatic disease, and remains clinically challenging breast cancer subtype to treat. Despite the survival dependency on the proteasome pathway genes, FDA-approved proteasome inhibitors induced minimal clinical response in breast cancer patients due to weak proteasome inhibition. Hence, developing effective targeted therapy using potent proteasome inhibitor is required. Methods: We evaluated anti-cancer activity of a potent proteasome inhibitor, marizomib, in vitro using breast cancer lines and in vivo using 4T1.2 murine syngeneic model, MDA-MB-231 xenografts, and patient-derived tumor xenografts. Global proteome profiling, western blots, and RT-qPCR were used to investigate the mechanism of action for marizomib. Effect of marizomib on lung and brain metastasis was evaluated using syngeneic 4T1BR4 murine TNBC model in vivo. Results: We show that marizomib inhibits multiple proteasome catalytic activities and induces a better anti-tumor response in TNBC cell lines and patient-derived xenografts alone and in combination with the standard-of-care chemotherapy. Mechanistically, we show that marizomib is a dual inhibitor of proteasome and oxidative phosphorylation (OXPHOS) in TNBCs. Marizomib reduces lung and brain metastases by reducing the number of circulating tumor cells and the expression of genes involved in the epithelial-to-mesenchymal transition. We demonstrate that marizomib-induced OXPHOS inhibition upregulates glycolysis to meet the energetic demands of TNBC cells and combined inhibition of glycolysis with marizomib leads to a synergistic anti-cancer activity. Conclusions: Our data provide a strong rationale for a clinical evaluation of marizomib in primary and metastatic TNBC patients.
Collapse
|
26
|
Rausch JL, Ali AA, Lee DM, Gebreyohannes YK, Mehalek KR, Agha A, Patil SS, Tolstov Y, Wellens J, Dhillon HS, Makielski KR, Debiec-Rychter M, Schöffski P, Wozniak A, Duensing A. Differential antitumor activity of compounds targeting the ubiquitin-proteasome machinery in gastrointestinal stromal tumor (GIST) cells. Sci Rep 2020; 10:5178. [PMID: 32198455 PMCID: PMC7083865 DOI: 10.1038/s41598-020-62088-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 03/03/2020] [Indexed: 11/09/2022] Open
Abstract
The majority of gastrointestinal stromal tumors (GISTs) are driven by oncogenic KIT signaling and can therefore be effectively treated with the tyrosine kinase inhibitor (TKI) imatinib mesylate. However, most GISTs develop imatinib resistance through secondary KIT mutations. The type of resistance mutation determines sensitivity to approved second-/third-line TKIs but shows high inter- and intratumoral heterogeneity. Therefore, therapeutic strategies that target KIT independently of the mutational status are intriguing. Inhibiting the ubiquitin-proteasome machinery with bortezomib is effective in GIST cells through a dual mechanism of KIT transcriptional downregulation and upregulation of the pro-apoptotic histone H2AX but clinically problematic due to the drug’s adverse effects. We therefore tested second-generation inhibitors of the 20S proteasome (delanzomib, carfilzomib and ixazomib) with better pharmacologic profiles as well as compounds targeting regulators of ubiquitination (b-AP15, MLN4924) for their effectiveness and mechanism of action in GIST. All three 20S proteasome inhibitors were highly effective in vitro and in vivo, including in imatinib-resistant models. In contrast, b-AP15 and MLN4924 were only effective at high concentrations or had mostly cytostatic effects, respectively. Our results confirm 20S proteasome inhibitors as promising strategy to overcome TKI resistance in GIST, while highlighting the complexity of the ubiquitin-proteasome machinery as a therapeutic target.
Collapse
Affiliation(s)
- Jessica L Rausch
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Areej A Ali
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Donna M Lee
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Yemarshet K Gebreyohannes
- Departments of Oncology and General Medical Oncology, University Hospitals Leuven and KU Leuven Cancer Institute, Leuven, Belgium
| | - Keith R Mehalek
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Aya Agha
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Sneha S Patil
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Yanis Tolstov
- Molecular Urooncology, University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Jasmien Wellens
- Departments of Oncology and General Medical Oncology, University Hospitals Leuven and KU Leuven Cancer Institute, Leuven, Belgium
| | - Harbir S Dhillon
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | | | - Maria Debiec-Rychter
- Department of Human Genetics, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Patrick Schöffski
- Departments of Oncology and General Medical Oncology, University Hospitals Leuven and KU Leuven Cancer Institute, Leuven, Belgium
| | - Agnieszka Wozniak
- Departments of Oncology and General Medical Oncology, University Hospitals Leuven and KU Leuven Cancer Institute, Leuven, Belgium
| | - Anette Duensing
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA. .,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
27
|
Biotinylated HPMA centered polymeric nanoparticles for Bortezomib delivery. Int J Pharm 2020; 579:119173. [PMID: 32097684 DOI: 10.1016/j.ijpharm.2020.119173] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/16/2020] [Accepted: 02/21/2020] [Indexed: 12/23/2022]
Abstract
Bortezomib (BTZ) is a proteasome inhibitor as approved by US FDA for the treatment of multiple myeloma. It exhibits significant anti-cancer properties, against solid tumors; but lacks aqueous solubility, chemical stability which hinders its successful formulation development. The present study is an attempt to deliver BTZ using N-(2-hydroxypropyl) methacrylamide (HPMA) based copolymeric conjugates and biotinylated PNPs in an effective manner. Study describes a systematic synthetic pathway to synthesize functional polymeric conjugates such as HPMA-Biotin (HP-BT) HPMA-Polylactic acid (HPLA) and HPMA-PLA-Biotin (HPLA-BT) followed by exhaustive characterization both spectroscopically and microscopically. Our strategy yielded polymeric nanoparticles (PNPs) of narrow size range of 199.7 ± 1.32 nm. Release studies were performed at pH 7.4 and 5.6. PNPs were 2-folds less hemolytic (p < 0.0001) than pure drug. BTZ loaded PNPs of HPLA-BT demonstrated significant anti-cancer activity against MCF-7 cells. IC50 value of these PNPs was 56.06 ± 0.12 nM, which was approximately two folds less than BTZ (p < 0.0001). Cellular uptake study confirmed that higher uptake of formulations might be an outcome of biotin surface tethering characteristics that enhanced selectivity and targeting of formulations efficiently. In vivo pharmacokinetics evidenced increased bioavailability (AUC0 t-∞) of DL-HPLA-BT PNPs (drug loaded) than BTZ with an improved half-life. Overall the developed PNPs led to the improved and effective BTZ delivery.
Collapse
|
28
|
Parasrampuria DA, He J, Zhang L, Muresan B, Hu P, Nemat S, Hashim M, Lam A, Appiani C, Cavo M, Dimopoulos MA, San-Miguel J, Mateos MV. Comparison of efficacy from two different dosing regimens of bortezomib: an exposure-response analysis. Br J Haematol 2020; 189:860-868. [PMID: 32068255 DOI: 10.1111/bjh.16446] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/08/2019] [Indexed: 11/30/2022]
Abstract
Bortezomib is a first-in-class proteasome inhibitor, approved for the treatment of multiple myeloma. The originally approved dosing schedule of bortezomib results in significant toxicities that require dose interruptions and discontinuations. Consequentially, less frequent dosing has been explored to optimise bortezomib's benefit-risk profile. Here, we performed exposure-response analysis to compare the efficacy of the original bortezomib dosing regimen with less frequent dosing of bortezomib over nine 6-week treatment cycles using data from the VISTA clinical trial and the control arm of the ALCYONE clinical trial. The relationship between cumulative bortezomib dose and clinical response was evaluated with a univariate logit model. The median cumulative bortezomib dose was higher in ALCYONE versus VISTA (42·2 vs. 38·5 mg/m2 ) and ALCYONE patients stayed on treatment longer (mean: 7·2 vs. 5·8 cycles). For all endpoints and regimens, probability of clinical response correlated with cumulative bortezomib dose. Similar to results observed for VISTA, overall survival was longer in ALCYONE patients with ≥ 39·0 versus < 39·0 mg/m2 cumulative dose (hazard ratio, 0·119; P < 0·0001). Less frequent bortezomib dosing results in comparable efficacy, and a higher cumulative dose than the originally approved bortezomib dosing schedule, which may be in part be due to reduced toxicity and fewer dose reductions/interruptions.
Collapse
Affiliation(s)
| | - Jianming He
- Janssen Global Services, LLC, Raritan, NJ, USA
| | - Liping Zhang
- Janssen Research & Development, LLC, Spring House, PA, USA
| | | | - Peter Hu
- Janssen Research and Development, LLC, Raritan, NJ, USA
| | | | | | - Annette Lam
- Janssen Global Services, LLC, Raritan, NJ, USA
| | | | - Michele Cavo
- Seràgnoli Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | | | - Jesus San-Miguel
- Clínica Universidad de Navarra-CIMA, IDISNA, CIBERONC, Pamplona, Spain
| | | |
Collapse
|
29
|
Zhang L, Mager DE. Population-based meta-analysis of bortezomib exposure-response relationships in multiple myeloma patients. J Pharmacokinet Pharmacodyn 2020; 47:77-90. [PMID: 31939004 DOI: 10.1007/s10928-019-09670-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 12/31/2019] [Indexed: 10/25/2022]
Abstract
Bortezomib (Velcade®) is a reversible proteasome inhibitor that shows potent antineoplastic activity, by inhibiting the constitutively increased proteasome activity in myeloma cells, and is approved as a first-line therapy for multiple myeloma (MM). Although clinically successful, bortezomib exhibits a relatively narrow therapeutic index and can induce dose-limiting toxicities such as thrombocytopenia. This study aims to develop a quantitative and predictive pharmacodynamic model to investigate bortezomib dosing-regimens in a rational and efficient manner. Mean temporal profiles of bortezomib pharmacokinetics, proteasome activity, M-protein concentrations, and platelet counts following bortezomib monotherapy were extracted from published clinical studies. A population-based meta-analysis of bortezomib anti-myeloma activity and thrombocytopenia was conducted sequentially with a Stochastic Approximation Expectation Maximization algorithm in Monolix. The final pharmacodynamic model integrates drug-target interactions and cell signaling dynamics with temporal biomarkers of clinical efficacy and toxicity. Bortezomib pharmacokinetics, disease progression, and platelet dynamic profiles were well characterized in MM patients, and a local sensitivity analysis of the final model suggests that increased proteasome concentration could ultimately attenuate bortezomib antineoplastic activity in MM patients. In addition, model simulations confirm that a once-weekly dosing schedule represents an optimal therapeutic regimen with comparable antineoplastic activity but significantly reduced risk of thrombocytopenia. In conclusion, a pharmacodynamic model was successfully developed, which provides a quantitative, mechanism-based platform for probing bortezomib dosing-regimens. Further research is needed to determine whether this model could be used to individualize bortezomib regimens to maximize antineoplastic efficacy and minimize thrombocytopenia during MM treatment.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University At Buffalo, State University of New York, Buffalo, NY, 14214, USA.
| |
Collapse
|
30
|
What the Intensivists Need to Know About Critically Ill Myeloma Patients. ONCOLOGIC CRITICAL CARE 2020. [PMCID: PMC7121630 DOI: 10.1007/978-3-319-74588-6_98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by an increase in aberrant plasma cells in the bone marrow leading to rising monoclonal protein in serum and urine. With the introduction of novel therapies with manageable side effects, this incurable disease has evolved into a chronic disease with an acceptable quality of life for the majority of patients. Accordingly, management of acute complications is fundamental in reducing the morbidity and mortality in MM. MM emergencies include symptoms and signs related directly to the disease and/or to the treatment; many organs may be involved including, but not limited to, renal, cardiovascular, neurologic, hematologic, and infectious complications. This review will focus on the numerous approaches that are aimed at managing these complications.
Collapse
|
31
|
Liu M, Tang X, Ding J, Liu M, Zhao B, Deng Y, Song Y. A Sialylated-Bortezomib Prodrug Strategy Based on a Highly Expressed Selectin Target for the Treatment of Leukemia or Solid Tumors. Pharm Res 2019; 36:176. [PMID: 31686241 DOI: 10.1007/s11095-019-2714-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE This study aimed to explore the potential of sialic acid - related selectin targeting strategy in the treatment of leukemia and some solid tumors. We expected it could "actively" bind tumor cells and kill them, reducing non-specific toxicity to normal cells. METHODS BOR-SA prodrug was synthesized by reacting an ortho-dihydroxy group in SA with a boronic acid group in BOR. Two kinds of leukemia cells (RAW264.7 and HL60 cells), one solid sarcoma cell model (S180 cells) and their corresponding normal cells (monocytes (MO), neutrophil (NE) and fibroblast (L929)) were selected for the in vitro cell experiments (cytotoxicity, cellular uptake, cell cycle and apoptosis experiments). The S180 tumor-bearing Kunming mice model was established for anti-tumor pharmacodynamic experiments. RESULTS In vitro cell assay results showed that uptake of BOR-SA by HL60 and S180 cells were increased compared with the control group. BOR-SA induced a lower IC50, higher ratio of apoptosis and cell cycle arrest of tumor cells. In vivo anti-S180 tumor pharmacodynamics experiments showed that mice in the BOR-SA group had higher tumor inhibition rate, higher body weight and lower immune organ toxicity compared with the control group. CONCLUSIONS sialic acid-mediated selectin targeting strategy may have great potential in the treatment of related tumors.
Collapse
Affiliation(s)
- Mingqi Liu
- College of Pharmacy, Shenyang Pharmaceutical University, 85 Hongliu Road, Benxi, 117004, Liaoning, China
| | - Xueying Tang
- College of Pharmacy, Shenyang Pharmaceutical University, 85 Hongliu Road, Benxi, 117004, Liaoning, China
| | - Junqiang Ding
- College of Pharmacy, Shenyang Pharmaceutical University, 85 Hongliu Road, Benxi, 117004, Liaoning, China
| | - Mengyang Liu
- College of Pharmacy, Shenyang Pharmaceutical University, 85 Hongliu Road, Benxi, 117004, Liaoning, China
| | - Bowen Zhao
- College of Pharmacy, Shenyang Pharmaceutical University, 85 Hongliu Road, Benxi, 117004, Liaoning, China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, 85 Hongliu Road, Benxi, 117004, Liaoning, China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, 85 Hongliu Road, Benxi, 117004, Liaoning, China.
| |
Collapse
|
32
|
Giallongo C, Tibullo D, Camiolo G, Parrinello NL, Romano A, Puglisi F, Barbato A, Conticello C, Lupo G, Anfuso CD, Lazzarino G, Li Volti G, Palumbo GA, Di Raimondo F. TLR4 signaling drives mesenchymal stromal cells commitment to promote tumor microenvironment transformation in multiple myeloma. Cell Death Dis 2019; 10:704. [PMID: 31541083 PMCID: PMC6754430 DOI: 10.1038/s41419-019-1959-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023]
Abstract
Inflammation represents a key feature and hallmark of tumor microenvironment playing a major role in the interaction with mesenchymal stromal cells (MSC) in cancer progression. The aim of the present study was to investigate the crosstalk between MSCs and myeloma cells (MM) in the pro-inflammatory microenvironment promoting immune evasion and tumor growth. MSC were collected from patients with diagnosis of MGUS (n = 10), smoldering myeloma (n = 7), multiple myeloma at diagnosis (n = 16), relapse (n = 5) or refractory (n = 3), and from age-matched healthy controls (HC, n = 10) and cultured with peripheral blood mononucleated cells (PBMC) from healthy volunteer donors. Similarly to MM, we showed that MSC from smoldering multiple myeloma (SMM) patients activated neutrophils and conferred an immunosuppressive and pro-angiogenic phenotype. Furthermore, co-cultures of plasma cells (PC) and HC-MSC suggested that such activation is driven by MM cells through the switching into a pro-inflammatory phenotype mediated by toll-like receptor 4 (TLR4). These results were further confirmed using a zebrafish as an immunocompetent in vivo model, showing the role of MM-MSC in supporting PCs engraftment and Th2 response. Such effect was abolished following inhibition of TLR4 signaling in MM-MSC before co-injection with PC. Moreover, the addition of a TLR4 inhibitor in the co-culture of HC-MSC with MM cells prevented the activation of the pro-tumor activity in PC-educated MSC. In conclusion, our study provides evidence that TLR4 signaling plays a key role in MSC transformation by inducing a pro-tumor phenotype associated with a permissive microenvironment allowing immune escape and tumor growth.
Collapse
Affiliation(s)
- Cesarina Giallongo
- Section of Haematology, Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
- EuroMediterranean Institute of Science and Technology, Palermo, Italy.
| | - Giuseppina Camiolo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nunziatina L Parrinello
- Section of Haematology, Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Alessandra Romano
- Section of Haematology, Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Fabrizio Puglisi
- Section of Haematology, Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Alessandro Barbato
- Section of Haematology, Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Concetta Conticello
- Section of Haematology, Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Gabriella Lupo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Giacomo Lazzarino
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, Largo F. Vito 1, 00168, Rome, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
- EuroMediterranean Institute of Science and Technology, Palermo, Italy.
| | - Giuseppe Alberto Palumbo
- Department of Medical, Surgical Sciences and Advanced Technologies "G. F. Ingrassia", University of Catania, Catania, Italy
| | - Francesco Di Raimondo
- Section of Haematology, Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| |
Collapse
|
33
|
Glycine-Poly-L-Lactic Acid Copolymeric Nanoparticles for the Efficient Delivery of Bortezomib. Pharm Res 2019; 36:160. [PMID: 31520196 DOI: 10.1007/s11095-019-2686-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/13/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Bortezomib (BTZ) is a proteasome inhibitor used for multiple myeloma and mantle cell lymphoma treatment. BTZ's aqueous in solubility is the main hindrance in its successful development as a commercial formulation. The main objective of the present study is to develop and characterize folic acid-glycine-poly-L-lactic acid (FA-Gly4-PLA) based nanoformulation (NPs) to improve solubility and efficacy of BTZ. METHODS BTZ loaded FA-Gly4-PLA NPs were prepared and characterized for size, zeta potential, in vitro studies such as release, kinetics modeling, hemolytic toxicity, and cell line-based studies (Reactive Oxygen Species: ROS and cytotoxicity). RESULTS BTZ loaded NPs (BTZ-loaded FA-Gly4-PLA) and blank NPs (FA-Gly4-PLA) size, zeta, and PDI were found to be 110 ± 8.1 nm, 13.7 ± 1.01 mV, 0.19 ± 0.03 and 198 ± 9.01 nm, 8.63 ± 0.21 mV, 0.21 ± 0.08 respectively. The percent encapsulation efficiency (% EE) and percent drug loading (% DL) of BTZ loaded FA-Gly4-PLA NPs was calculated to be 78.3 ± 4.1 and 12.38 ± 2.1. The Scanning Electron Microscopy (SEM) showed that NPs were slightly biconcave in shape. The in vitro release of BTZ from FA-Gly4-PLA NPs resulted in the sustained manner. The prepared NPs were less hemolytic than BTZ. CONCLUSIONS BTZ loaded Gly4-PLA NPs apoptotic index was found to be much higher than BTZ but lesser than BTZ loaded FA-Gly4-PLA against breast cancer cell lines (MDA-MB-231). ROS intracellular assessment assay indicated that BTZ and BTZ loaded FA-Gly4-PLA NPs exhibited higher ROS production. Conclusively, the BTZ loaded FA-Gly4-PLA NPs were able to encapsulate more BTZ than BTZ loaded Gly4-PLA NPs and were found to be more effective as per as in vitro anti-cancer effect is concerned.
Collapse
|
34
|
Blosser N, Jupp J, Yau P, Stewart D. Clinical Pharmacokinetic and Pharmacodynamic Considerations in Treating Non-Hodgkin Lymphoma. Clin Pharmacokinet 2019; 59:7-23. [PMID: 31385204 DOI: 10.1007/s40262-019-00807-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Non-Hodgkin lymphoma (NHL) includes a variety of closely related malignancies that originate from lymphoid precursors. The majority of NHLs are of B-cell lineage, for which traditional therapy involves chemotherapy in combination with the anti-CD20 monoclonal antibody rituximab. Ongoing research into the pathogenesis of NHL subtypes has given rise to the use of novel agents that target specific molecular pathways. While the incidence of NHL extends over a range of ages from pediatric to elderly settings, the majority of diagnoses occur over age 60 years. Increasing the use of concomitant medication coupled with declining organ function among this group of patients creates pharmacokinetic (PK) challenges in administering a number of agents involved in the treatment of NHL. In addition, since many of the new agents are administered orally, there are a number of added PK factors that must be taken into consideration with their prescribing and administration. This article will review the available literature on the PK and pharmacodynamic properties of agents commonly used in the treatment of NHL, and intends to provide information that can assist with properly using these drugs in this setting.
Collapse
Affiliation(s)
- Nikki Blosser
- Pharmacy Services, Tom Baker Cancer Centre, Alberta Health Services, Edmonton, AB, Canada
| | - Jennifer Jupp
- Pharmacy Services, Tom Baker Cancer Centre, Alberta Health Services, Edmonton, AB, Canada
| | - Patrick Yau
- Pharmacy Services, Tom Baker Cancer Centre, Alberta Health Services, Edmonton, AB, Canada
| | - Douglas Stewart
- Departments of Oncology and Medicine, University of Calgary and Tom Baker Cancer Centre, Alberta Health Services, 1331-29th Street NW, Calgary, AB, T2N4N2, Canada.
| |
Collapse
|
35
|
Grosset AA, Ouellet V, Caron C, Fragoso G, Barrès V, Delvoye N, Latour M, Aprikian A, Bergeron A, Chevalier S, Fazli L, Fleshner N, Gleave M, Karakiewicz P, Lacombe L, Lattouf JB, van der Kwast T, Trudel D, Mes-Masson AM, Saad F. Validation of the prognostic value of NF-κB p65 in prostate cancer: A retrospective study using a large multi-institutional cohort of the Canadian Prostate Cancer Biomarker Network. PLoS Med 2019; 16:e1002847. [PMID: 31265453 PMCID: PMC6605640 DOI: 10.1371/journal.pmed.1002847] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 06/03/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The identification of patients with high-risk prostate cancer (PC) is a major challenge for clinicians, and the improvement of current prognostic parameters is an unmet clinical need. We and others have identified an association between the nuclear localization of NF-κB p65 and biochemical recurrence (BCR) in PC in small and/or single-centre cohorts of patients. METHODS AND FINDINGS In this study, we accessed 2 different multi-centre tissue microarrays (TMAs) representing cohorts of patients (Test-TMA and Validation-TMA series) of the Canadian Prostate Cancer Biomarker Network (CPCBN) to validate the association between p65 nuclear frequency and PC outcomes. Immunohistochemical staining of p65 was performed on the Test-TMA and Validation-TMA series, which include PC tissues from patients treated by first-line radical prostatectomy (n = 250 and n = 1,262, respectively). Two independent observers evaluated the p65 nuclear frequency in digital images of cancer tissue and benign adjacent gland tissue. Kaplan-Meier curves coupled with a log-rank test and univariate and multivariate Cox regression models were used for statistical analyses of continuous values and dichotomized data (cutoff of 3%). Multivariate analysis of the Validation-TMA cohort showed that p65 nuclear frequency in cancer cells was an independent predictor of BCR using continuous (hazard ratio [HR] 1.02 [95% CI 1.00-1.03], p = 0.004) and dichotomized data (HR 1.33 [95% CI 1.09-1.62], p = 0.005). Using a cutoff of 3%, we found that this biomarker was also associated with the development of bone metastases (HR 1.82 [95% CI 1.05-3.16], p = 0.033) and PC-specific mortality (HR 2.63 [95% CI 1.30-5.31], p = 0.004), independent of clinical parameters. BCR-free survival, bone-metastasis-free survival, and PC-specific survival were shorter for patients with higher p65 nuclear frequency (p < 0.005). As the small cores on TMAs are a limitation of the study, a backward validation of whole PC tissue section will be necessary for the implementation of p65 nuclear frequency as a PC biomarker in the clinical workflow. CONCLUSIONS We report the first study using the pan-Canadian multi-centre cohorts of CPCBN and validate the association between increased frequency of nuclear p65 frequency and a risk of disease progression.
Collapse
Affiliation(s)
- Andrée-Anne Grosset
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Véronique Ouellet
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Christine Caron
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Gabriela Fragoso
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Véronique Barrès
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Nathalie Delvoye
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Mathieu Latour
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Armen Aprikian
- Research Institute of the McGill University Health Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Alain Bergeron
- Axe Oncologie, Centre de recherche du Centre hospitalier universitaire de Québec–Université Laval, Centre hospitalier universitaire de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de recherche sur le cancer, Université Laval, Quebec, Quebec, Canada
- Département de chirurgie, Université Laval, Quebec, Quebec, Canada
| | - Simone Chevalier
- Research Institute of the McGill University Health Centre, McGill University Health Centre, Montreal, Quebec, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | | | - Martin Gleave
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Pierre Karakiewicz
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Louis Lacombe
- Axe Oncologie, Centre de recherche du Centre hospitalier universitaire de Québec–Université Laval, Centre hospitalier universitaire de Québec–Université Laval, Quebec, Quebec, Canada
- Centre de recherche sur le cancer, Université Laval, Quebec, Quebec, Canada
- Département de chirurgie, Université Laval, Quebec, Quebec, Canada
| | - Jean-Baptiste Lattouf
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | | | - Dominique Trudel
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| | - Fred Saad
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
- * E-mail:
| | | |
Collapse
|
36
|
Baljevic M, Orlowski RZ. Pharmacodynamics and pharmacokinetics of proteasome inhibitors for the treatment of multiple myeloma. Expert Opin Drug Metab Toxicol 2019; 15:459-473. [PMID: 31104525 PMCID: PMC10393465 DOI: 10.1080/17425255.2019.1621839] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 05/17/2019] [Indexed: 12/18/2022]
Abstract
Introduction: Multiple myeloma (MM) is the second most commonly diagnosed hematologic malignancy and has an increasing incidence and prevalence globally, and proteasome inhibitors (PIs) form the backbone of some of our most effective regimens for all phases of this disease in fit and frail patients. Areas covered: Strong understanding of the proteasome complex is increasingly important as the rapid development of new PIs and innovative myeloma therapies complicate the use of old and new combination regimens. We focus herein on the pharmacodynamics and pharmacokinetics of the approved PIs and others in development, including their safety and efficacy in corresponding clinical studies. Expert opinion: Advancements such as the first oral PI, ixazomib, with a more convenient route of administration and improved toxicity profile led to an improved quality of life, patient compliance, and all-oral combination regimens which are efficacious for long-term management of standard and high-risk MM. Novel pan-PIs, such as marizomib, hold the promise of superior clinical activity due to irreversible targeting of all multicatalytic proteinase complex subunits. Development of clinically validated biomarkers of PI sensitivity/resistance is required to inform utilization of the most optimal and effective, rationally targeted PI treatments for all MM patients.
Collapse
Affiliation(s)
- Muhamed Baljevic
- The University of Nebraska Medical Center, Division of Hematology and Oncology, Omaha, NE, USA
| | - Robert Z. Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
37
|
Schneider SM, Pritchard SM, Wudiri GA, Trammell CE, Nicola AV. Early Steps in Herpes Simplex Virus Infection Blocked by a Proteasome Inhibitor. mBio 2019; 10:e00732-19. [PMID: 31088925 PMCID: PMC6520451 DOI: 10.1128/mbio.00732-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/08/2019] [Indexed: 12/17/2022] Open
Abstract
Viruses commandeer host cell 26S proteasome activity to promote viral entry, gene expression, replication, assembly, and egress. Proteasomal degradation activity is critical for herpes simplex virus (HSV) infection. The proteasome inhibitor bortezomib (also known as Velcade and PS-341) is a clinically effective antineoplastic drug that is FDA approved for treatment of hematologic malignancies such as multiple myeloma and mantle cell lymphoma. Low nanomolar concentrations of bortezomib inhibited infection by HSV-1, HSV-2, and acyclovir-resistant strains. Inhibition coincided with minimal cytotoxicity. Bortezomib did not affect attachment of HSV to cells or inactivate the virus directly. Bortezomib acted early in HSV infection by perturbing two distinct proteasome-dependent steps that occur within the initial hours of infection: the transport of incoming viral nucleocapsids to the nucleus and the virus-induced disruption of host nuclear domain 10 (ND10) structures. The combination of bortezomib with acyclovir demonstrated synergistic inhibitory effects on HSV infection. Thus, bortezomib is a novel potential therapeutic for HSV with a defined mechanism of action.IMPORTANCE Viruses usurp host cell functions to advance their replicative agenda. HSV relies on cellular proteasome activity for successful infection. Proteasome inhibitors, such as MG132, block HSV infection at multiple stages of the infectious cycle. Targeting host cell processes for antiviral intervention is an unconventional approach that might limit antiviral resistance. Here we demonstrated that the proteasome inhibitor bortezomib, which is a clinically effective cancer drug, has the in vitro features of a promising anti-HSV therapeutic. Bortezomib inhibited HSV infection during the first hours of infection at nanomolar concentrations that were minimally cytotoxic. The mechanism of bortezomib's inhibition of early HSV infection was to halt nucleocapsid transport to the nucleus and to stabilize the ND10 cellular defense complex. Bortezomib and acyclovir acted synergistically to inhibit HSV infection. Overall, we present evidence for the repurposing of bortezomib as a novel antiherpesviral agent and describe specific mechanisms of action.
Collapse
Affiliation(s)
- Seth M Schneider
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Suzanne M Pritchard
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - George A Wudiri
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Chasity E Trammell
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Anthony V Nicola
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
38
|
Chong PY, Shotwell JB, Miller J, Price DJ, Maynard A, Voitenleitner C, Mathis A, Williams S, Pouliot JJ, Creech K, Wang F, Fang J, Zhang H, Tai VWF, Turner E, Kahler KM, Crosby R, Peat AJ. Design of N-Benzoxaborole Benzofuran GSK8175-Optimization of Human Pharmacokinetics Inspired by Metabolites of a Failed Clinical HCV Inhibitor. J Med Chem 2019; 62:3254-3267. [PMID: 30763090 PMCID: PMC6466479 DOI: 10.1021/acs.jmedchem.8b01719] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
![]()
We previously described the discovery
of GSK5852 (1), a non-nucleoside polymerase (NS5B) inhibitor
of hepatitis C virus (HCV), in which an N-benzyl
boronic acid was essential for potent antiviral activity. Unfortunately,
facile benzylic oxidation resulted in a short plasma half-life (5
h) in human volunteers, and a backup program was initiated to remove
metabolic liabilities associated with 1. Herein, we describe
second-generation NS5B inhibitors including GSK8175 (49), a sulfonamide-N-benzoxaborole analog with low
in vivo clearance across preclinical species and broad-spectrum activity
against HCV replicons. An X-ray structure of NS5B protein cocrystallized
with 49 revealed unique protein-inhibitor interactions
mediated by an extensive network of ordered water molecules and the
first evidence of boronate complex formation within the binding pocket.
In clinical studies, 49 displayed a 60–63 h half-life
and a robust decrease in viral RNA levels in HCV-infected patients,
thereby validating our hypothesis that reducing benzylic oxidation
would improve human pharmacokinetics and lower efficacious doses relative
to 1.
Collapse
Affiliation(s)
- Pek Y Chong
- GlaxoSmithKline , 5 Moore Drive , Research Triangle Park , North Carolina 27709 , United States
| | - J Brad Shotwell
- GlaxoSmithKline , 5 Moore Drive , Research Triangle Park , North Carolina 27709 , United States
| | - John Miller
- GlaxoSmithKline , 5 Moore Drive , Research Triangle Park , North Carolina 27709 , United States
| | - Daniel J Price
- GlaxoSmithKline , 200 Cambridge Park Drive , Cambridge , Massachusetts 02140 , United States
| | - Andy Maynard
- GlaxoSmithKline , 5 Moore Drive , Research Triangle Park , North Carolina 27709 , United States
| | - Christian Voitenleitner
- GlaxoSmithKline , 5 Moore Drive , Research Triangle Park , North Carolina 27709 , United States
| | - Amanda Mathis
- GlaxoSmithKline , 5 Moore Drive , Research Triangle Park , North Carolina 27709 , United States
| | - Shawn Williams
- GlaxoSmithKline , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Jeffrey J Pouliot
- GlaxoSmithKline , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Katrina Creech
- GlaxoSmithKline , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Feng Wang
- GlaxoSmithKline , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Jing Fang
- GlaxoSmithKline , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Huichang Zhang
- GlaxoSmithKline , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Vincent W-F Tai
- GlaxoSmithKline , 5 Moore Drive , Research Triangle Park , North Carolina 27709 , United States
| | - Elizabeth Turner
- GlaxoSmithKline , 5 Moore Drive , Research Triangle Park , North Carolina 27709 , United States
| | - Kirsten M Kahler
- GlaxoSmithKline , 5 Moore Drive , Research Triangle Park , North Carolina 27709 , United States
| | - Renae Crosby
- GlaxoSmithKline , 5 Moore Drive , Research Triangle Park , North Carolina 27709 , United States
| | - Andrew J Peat
- GlaxoSmithKline , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| |
Collapse
|
39
|
Pastorino F, Brignole C, Di Paolo D, Perri P, Curnis F, Corti A, Ponzoni M. Overcoming Biological Barriers in Neuroblastoma Therapy: The Vascular Targeting Approach with Liposomal Drug Nanocarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1804591. [PMID: 30706636 DOI: 10.1002/smll.201804591] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/22/2018] [Indexed: 06/09/2023]
Abstract
Neuroblastoma is a rare pediatric cancer characterized by a wide clinical behavior and adverse outcome despite aggressive therapies. New approaches based on targeted drug delivery may improve efficacy and decrease toxicity of cancer therapy. Furthermore, nanotechnology offers additional potential developments for cancer imaging, diagnosis, and treatment. Following these lines, in the past years, innovative therapies based on the use of liposomes loaded with anticancer agents and functionalized with peptides capable of recognizing neuroblastoma cells and/or tumor-associated endothelial cells have been developed. Studies performed in experimental orthotopic models of human neuroblastoma have shown that targeted nanocarriers can be exploited for not only decreasing the systemic toxicity of the encapsulated anticancer drugs, but also increasing their tumor homing properties, enhancing tumor vascular permeability and perfusion (and, consequently, drug penetration), inducing tumor apoptosis, inhibiting angiogenesis, and reducing tumor glucose consumption. Furthermore, peptide-tagged liposomal formulations are proved to be more efficacious in inhibiting tumor growth and metastatic spreading of neuroblastoma than nontargeted liposomes. These findings, herein reviewed, pave the way for the design of novel targeted liposomal nanocarriers useful for multitargeting treatment of neuroblastoma.
Collapse
Affiliation(s)
- Fabio Pastorino
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Chiara Brignole
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Daniela Di Paolo
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Patrizia Perri
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| | - Flavio Curnis
- Tumor Biology and Vascular Targeting Unit, IRCCS San Raffaele Scientific Institute, 16132, Milan, Italy
| | - Angelo Corti
- Tumor Biology and Vascular Targeting Unit, IRCCS San Raffaele Scientific Institute, 16132, Milan, Italy
- Vita Salute San Raffaele University, 16132, Milan, Italy
| | - Mirco Ponzoni
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy
| |
Collapse
|
40
|
Bastola P, Oien DB, Cooley M, Chien J. Emerging Cancer Therapeutic Targets in Protein Homeostasis. AAPS JOURNAL 2018; 20:94. [PMID: 30151644 DOI: 10.1208/s12248-018-0254-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/10/2018] [Indexed: 12/11/2022]
Abstract
Genomic aberrations inside malignant cells through copy number alterations, aneuploidy, and mutations can exacerbate misfolded and unfolded protein burden resulting in increased proteotoxic stress. Increased proteotoxic stress can be deleterious to malignant cells; therefore, these cells rely heavily on the protein quality control mechanisms for survival and proliferation. Components of the protein quality control, such as the unfolded protein response, heat shock proteins, autophagy, and the ubiquitin proteasome system, orchestrate a cascade of downstream events that allow the mitigation of the proteotoxic stress. This dependency makes components of the protein quality control mechanisms attractive targets in cancer therapeutics. In this review, we explore the components of the protein homeostasis especially focusing on the emerging cancer therapeutic agents/targets that are being actively pursued actively.
Collapse
Affiliation(s)
- Prabhakar Bastola
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM, 87131, USA.,Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66130, USA
| | - Derek B Oien
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM, 87131, USA
| | - Megan Cooley
- Methods Development, Small Molecules, PRA Health Sciences, Lenexa, KS, 66215, USA
| | - Jeremy Chien
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud NE, Albuquerque, NM, 87131, USA.
| |
Collapse
|
41
|
Park JE, Miller Z, Jun Y, Lee W, Kim KB. Next-generation proteasome inhibitors for cancer therapy. Transl Res 2018; 198:1-16. [PMID: 29654740 PMCID: PMC6151281 DOI: 10.1016/j.trsl.2018.03.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/16/2018] [Accepted: 03/18/2018] [Indexed: 02/06/2023]
Abstract
Over 2 decades ago, the proteasome was considered a risky or even untenable therapeutic target. Today, proteasome inhibitors are a mainstay in the treatment of multiple myeloma (MM) and have sales in excess of 3 billion US dollars annually. More importantly, the availability of proteasome inhibitors has greatly improved the survival and quality of life for patients with MM. Despite the remarkable success of proteasome inhibitor therapies to date, the potential for improvement remains, and the development and optimal use of proteasome inhibitors as anticancer agents continues to be an active area of research. In this review, we briefly discuss the features and limitations of the 3 proteasome inhibitor drugs currently used in the clinic and provide an update on current efforts to develop next-generation proteasome inhibitors with the potential to overcome the limitations of existing proteasome inhibitor drugs.
Collapse
Affiliation(s)
- Ji Eun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Zachary Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky
| | - Yearin Jun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky.
| |
Collapse
|
42
|
Liu S, Ono RJ, Yang C, Gao S, Ming Tan JY, Hedrick JL, Yang YY. Dual pH-Responsive Shell-Cleavable Polycarbonate Micellar Nanoparticles for in Vivo Anticancer Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:19355-19364. [PMID: 29757607 DOI: 10.1021/acsami.8b01954] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
To exploit tumor and intracellular microenvironments, pH-responsive diblock copolymers of poly(ethylene glycol) and catechol-functionalized polycarbonate with acid-labile acetal bond as the linker are synthesized to prepare micellar nanoparticles that shed the shell at acidic tumor tissues and inside cancer cells, hence accelerating drug release at the target. The pH-dependent cleavage of the shell is demonstrated at pH 5.0 and 6.5 using 1H NMR. Bortezomib (BTZ, an anticancer drug containing a phenylboronic acid group) is conjugated to the polymers through formation of pH-responsive boronate ester bond between boronic acid and catechol in the polymers. Dual pH-responsive bortezomib-polymer conjugates (BTZ-PC) self-assemble into micellar nanoparticles of small size (<110 nm) with narrow size distribution and high drug loading capacity. Acidic pH accelerates BTZ release from BTZ-PC micelles and enhances intracelluar uptake of the micelles, hence increasing in vitro cytotoxicity against human breast cancer cells. More importantly, the BTZ-PC micelles achieve a stronger antitumor effect in a human breast cancer BT-474 xenograft mouse model than free BTZ and mitigate in vivo hepatotoxicity of BTZ. These dual pH-responsive shell-cleavable nanoparticles are a potentially promising carrier for BTZ delivery.
Collapse
Affiliation(s)
- Shaoqiong Liu
- Institute of Bioengineering and Nanotechnology , 31 Biopolis Way, The Nanos , 138669 , Singapore
| | - Robert J Ono
- IBM Almaden Research Center , 650 Harry Road , San Jose , California 95120 , United States
| | - Chuan Yang
- Institute of Bioengineering and Nanotechnology , 31 Biopolis Way, The Nanos , 138669 , Singapore
| | - Shujun Gao
- Institute of Bioengineering and Nanotechnology , 31 Biopolis Way, The Nanos , 138669 , Singapore
| | - Jordan Yong Ming Tan
- Institute of Bioengineering and Nanotechnology , 31 Biopolis Way, The Nanos , 138669 , Singapore
| | - James L Hedrick
- IBM Almaden Research Center , 650 Harry Road , San Jose , California 95120 , United States
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology , 31 Biopolis Way, The Nanos , 138669 , Singapore
| |
Collapse
|
43
|
HIV Protease-Generated Casp8p41, When Bound and Inactivated by Bcl2, Is Degraded by the Proteasome. J Virol 2018; 92:JVI.00037-18. [PMID: 29643240 PMCID: PMC6002723 DOI: 10.1128/jvi.00037-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 04/02/2018] [Indexed: 11/20/2022] Open
Abstract
HIV protease is known to cause cell death, which is dependent upon cleavage of procaspase 8. HIV protease cleavage of procaspase 8 generates Casp8p41, which directly binds Bak with nanomolar affinity, causing Bak activation and consequent cell death. Casp8p41 can also bind Bcl2 with nanomolar affinity, in which case cell death is averted. Central memory CD4 T cells express high levels of Bcl2, possibly explaining why those cells do not die when they reactivate HIV. Here, we determine that the Casp8p41-Bcl2 complex is polyubiquitinated and degraded by the proteasome. Ixazomib, a proteasome inhibitor in clinical use, blocks this pathway, increasing the abundance of Casp8p41 and causing more cells to die in a Casp8p41-dependent manner. IMPORTANCE The Casp8p41 pathway of cell death is unique to HIV-infected cells yet is blocked by Bcl2. Once bound by Bcl2, Casp8p41 is polyubiquitinated and degraded by the proteasome. Proteasome inhibition blocks degradation of Casp8p41, increasing Casp8p41 levels and causing more HIV-infected cells to die.
Collapse
|
44
|
Kaspers GJL, Niewerth D, Wilhelm BAJ, Scholte-van Houtem P, Lopez-Yurda M, Berkhof J, Cloos J, de Haas V, Mathôt RA, Attarbaschi A, Baruchel A, de Bont ES, Fagioli F, Rössig C, Klingebiel T, De Moerloose B, Nelken B, Palumbo G, Reinhardt D, Rohrlich PS, Simon P, von Stackelberg A, Zwaan CM. An effective modestly intensive re-induction regimen with bortezomib in relapsed or refractory paediatric acute lymphoblastic leukaemia. Br J Haematol 2018; 181:523-527. [PMID: 29676440 DOI: 10.1111/bjh.15233] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/16/2018] [Indexed: 01/30/2023]
Abstract
This trial explored the efficacy of re-induction chemotherapy including bortezomib in paediatric relapsed/refractory acute lymphoblastic leukaemia. Patients were randomized 1:1 to bortezomib (1.3 mg/m2 /dose) administered early or late to a dexamethasone and vincristine backbone. Both groups did not differ regarding peripheral blast count on day 8, the primary endpoint. After cycle 1, 8 of 25 (32%) patients achieved complete remission with incomplete blood count recovery, 7 (28%) a partial remission and 10 had treatment failure. Most common grade 3-4 toxicities were febrile neutropenia (31%) and pain (17%). Bortezomib was safely combined with vincristine. Bortezomib rarely penetrated the cerebrospinal fluid.
Collapse
Affiliation(s)
- Gertjan J L Kaspers
- Department of Paediatric Oncology/Haematology, VU University Medical Centre, Amsterdam, the Netherlands.,Princess Máxima Centre for Paediatric Oncology, Utrecht, the Netherlands
| | - Denise Niewerth
- Department of Paediatric Oncology/Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | - Bram A J Wilhelm
- Clinical Pharmacology and Pharmacy, VU University Medical Centre, Amsterdam, the Netherlands
| | - Peggy Scholte-van Houtem
- Paediatric Oncology, Erasmus MC, Rotterdam, the Netherlands.,Innovative Therapies for Children with Cancer Consortium, Paris, France
| | - Marta Lopez-Yurda
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, the Netherlands.,Paediatric Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Johannes Berkhof
- Department of Epidemiology & Biostatistics, VU University Medical Centre, Amsterdam, the Netherlands
| | - Jacqueline Cloos
- Department of Paediatric Oncology/Haematology, VU University Medical Centre, Amsterdam, the Netherlands
| | | | - Ron A Mathôt
- Clinical Pharmacology, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Andishe Attarbaschi
- Department of Paediatric Haematology and Oncology, St. Anna Children's Hospital, Department of Paediatrics and Adolescent Medicine, Medical University Vienna, Vienna, Austria
| | - André Baruchel
- Innovative Therapies for Children with Cancer Consortium, Paris, France.,Dept. of Paediatric Haematology, Hopital Saint Louis, Paris, France
| | - Eveline S de Bont
- Department of Paediatric Oncology/Haematology, Beatrix Children's Hospital, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Claudia Rössig
- Paediatric Haematology and Oncology, University Hospital Münster, Münster, Germany
| | - Thomas Klingebiel
- Department of Paediatrics, University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Brigitte Nelken
- Paediatric Haematology, Hospital Jeanne de Flandre, Lille, France
| | | | - Dirk Reinhardt
- Innovative Therapies for Children with Cancer Consortium, Paris, France.,AML-BFM Study Group, Paediatric Haematology/Oncology, University Children's Hospital Essen, Essen, Germany
| | | | - Pauline Simon
- Paediatric Oncology, University Hospital of Besancon, Besancon, France
| | - Arend von Stackelberg
- Department of Paediatric Oncology/Haematology, CharitéUniversitätsmedizin, Berlin, Germany
| | - Christian Michel Zwaan
- Paediatric Oncology, Erasmus MC, Rotterdam, the Netherlands.,Innovative Therapies for Children with Cancer Consortium, Paris, France
| |
Collapse
|
45
|
|
46
|
Raghu D, Paul PJ, Gulati T, Deb S, Khoo C, Russo A, Gallo E, Blandino G, Chan AL, Takano E, Sandhu SK, Fox SB, Williams S, Haupt S, Gamell C, Haupt Y. E6AP promotes prostate cancer by reducing p27 expression. Oncotarget 2018; 8:42939-42948. [PMID: 28477016 PMCID: PMC5522117 DOI: 10.18632/oncotarget.17224] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/16/2017] [Indexed: 12/03/2022] Open
Abstract
Prostate cancer (PC) is the most common cancer in men. Elevated levels of E3 ligase, E6-Associated Protein (E6AP) were previously linked to PC, consistent with increased protein expression in a subset of PC patients. In cancers, irregular E3 ligase activity drives proteasomal degradation of tumor suppressor proteins. Accordingly, E3 ligase inhibitors define a rational therapy to restore tumor suppression. The relevant tumor suppressors targeted by E6AP in PC are yet to be fully identified. In this study we show that p27, a key cell cycle regulator, is a target of E6AP in PC. Down regulation of E6AP increases p27 expression and enhances its nuclear accumulation in PC. We demonstrate that E6AP regulates p27 expression by inhibiting its transcription in an E2F1-dependent manner. Concomitant knockdown of E6AP and p27 partially restores PC cell growth, supporting the contribution of p27 to the overall effect of E6AP on prostate tumorigenesis. Overall, we unravelled the E6AP-p27 axis as a new promoter of PC, exposing an attractive target for therapy through the restoration of tumor suppression.
Collapse
Affiliation(s)
- Dinesh Raghu
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Piotr Jan Paul
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Twishi Gulati
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Siddhartha Deb
- Anatpath Services Pty Ltd, Gardenvale, Victoria, Australia
| | - Christine Khoo
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Andrea Russo
- Department of Surgical Pathology, Regina Elena Cancer Institute, Rome, Italy
| | - Enzo Gallo
- Department of Surgical Pathology, Regina Elena Cancer Institute, Rome, Italy
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, Italian National Cancer Institute, Rome, Italy
| | - Ai-Leen Chan
- Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Current address: Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| | - Elena Takano
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Shahneen K Sandhu
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Scott Williams
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Sue Haupt
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Cristina Gamell
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ygal Haupt
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Tumor Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia.,Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
47
|
Iannaccone A, Bruno G, Ravera A, Gay F, Salvini M, Bringhen S, Sabia L, Avenatti E, Veglio F, Milan A. Evaluation of Cardiovascular Toxicity Associated with Treatments Containing Proteasome Inhibitors in Multiple Myeloma Therapy. High Blood Press Cardiovasc Prev 2018; 25:209-218. [PMID: 29582365 DOI: 10.1007/s40292-018-0256-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/19/2018] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Recently new treatment options have substantially increased survival for patients with relapsed and/or refractory multiple myeloma (RRMM). Among these, proteasome inhibitors (PI), such as bortezomib and carfilzomib, offer high response rate and prolonged survival. These agents are generally well tolerated but demonstrated a significant cardiovascular toxicity, mostly for regimen containing carfilzomib. AIM To assess the cardiovascular damage in patients treated with PI for RRMM. METHODS 28 consecutive subjects treated with PI for RRMM were evaluated and compared with a population of 22 control (Con) subjects, matched for age, sex and mean 24 h blood pressure (24hMBP). All individuals underwent trans-thoracic echocardiography, ambulatory blood pressure monitoring and pulse wave velocity (PVW) study. RESULTS PI patients did not have significant differences in blood pressure load and PWV compared to controls. Among echocardiographic parameters, the global longitudinal strain (GLS) was significantly decreased in PI subjects (p = 0.02). The GLS was significantly lower also considering only patients treated with carfilzomib. Moreover, among carfilzomib patients, we found increase values of left ventricle mass indexed by BSA (LVMi; p = 0.047). After correction for age, sex, BSA, 24hMBP and morphological and functional parameters of LV, treatment with PI and carfilzomib were significantly associated with GLS (p = 0.01; p = 0.036, respectively). CONCLUSIONS PI treatment is associated with subclinical LV dysfunction in patients with RRMM compared to controls, as demonstrated by lower GLS values. These results are confirmed also considering patients treated with carfilzomib. Moreover, in this subgroup of patients, the LVMi is also increased, suggesting higher cardiotoxicity with this treatment.
Collapse
Affiliation(s)
- Andrea Iannaccone
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy.
| | - G Bruno
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| | - A Ravera
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| | - F Gay
- Myeloma Unit, Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University Hospital 'Città della Salute e della Scienza di Torino', University of Torino, Turin, Italy
| | - M Salvini
- Myeloma Unit, Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University Hospital 'Città della Salute e della Scienza di Torino', University of Torino, Turin, Italy
| | - S Bringhen
- Myeloma Unit, Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University Hospital 'Città della Salute e della Scienza di Torino', University of Torino, Turin, Italy
| | - L Sabia
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| | - E Avenatti
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| | - F Veglio
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| | - A Milan
- Hypertension Unit, Division of Internal Medicine, Department of Medical Sciences, University Hospital "Città della Salute e della Scienza", University of Torino, Turin, Italy
| |
Collapse
|
48
|
Allen JE, Kline CLB, Prabhu VV, Wagner J, Ishizawa J, Madhukar N, Lev A, Baumeister M, Zhou L, Lulla A, Stogniew M, Schalop L, Benes C, Kaufman HL, Pottorf RS, Nallaganchu BR, Olson GL, Al-Mulla F, Duvic M, Wu GS, Dicker DT, Talekar MK, Lim B, Elemento O, Oster W, Bertino J, Flaherty K, Wang ML, Borthakur G, Andreeff M, Stein M, El-Deiry WS. Discovery and clinical introduction of first-in-class imipridone ONC201. Oncotarget 2018; 7:74380-74392. [PMID: 27602582 PMCID: PMC5342060 DOI: 10.18632/oncotarget.11814] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 08/30/2016] [Indexed: 12/20/2022] Open
Abstract
ONC201 is the founding member of a novel class of anti-cancer compounds called imipridones that is currently in Phase II clinical trials in multiple advanced cancers. Since the discovery of ONC201 as a p53-independent inducer of TRAIL gene transcription, preclinical studies have determined that ONC201 has anti-proliferative and pro-apoptotic effects against a broad range of tumor cells but not normal cells. The mechanism of action of ONC201 involves engagement of PERK-independent activation of the integrated stress response, leading to tumor upregulation of DR5 and dual Akt/ERK inactivation, and consequent Foxo3a activation leading to upregulation of the death ligand TRAIL. ONC201 is orally active with infrequent dosing in animals models, causes sustained pharmacodynamic effects, and is not genotoxic. The first-in-human clinical trial of ONC201 in advanced aggressive refractory solid tumors confirmed that ONC201 is exceptionally well-tolerated and established the recommended phase II dose of 625 mg administered orally every three weeks defined by drug exposure comparable to efficacious levels in preclinical models. Clinical trials are evaluating the single agent efficacy of ONC201 in multiple solid tumors and hematological malignancies and exploring alternative dosing regimens. In addition, chemical analogs that have shown promise in other oncology indications are in pre-clinical development. In summary, the imipridone family that comprises ONC201 and its chemical analogs represent a new class of anti-cancer therapy with a unique mechanism of action being translated in ongoing clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Jo Ishizawa
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Avital Lev
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - Lanlan Zhou
- Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | | | | - Cyril Benes
- Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | | | | | | | - Gary L Olson
- Provid Pharmaceuticals, Monmouth Junction, NJ, USA
| | | | - Madeleine Duvic
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Mala K Talekar
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Bora Lim
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Joseph Bertino
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Keith Flaherty
- Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Michael L Wang
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Mark Stein
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | | |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Gastrointestinal toxicities are commonly reported following treatment with proteasome inhibitors. The first-generation proteasome inhibitor, bortezomib, induces significant gastrointestinal side effects including nausea, vomiting, diarrhoea, and constipation, occurring in up to 84% of patients. Despite the development of safer proteasome inhibitors, such as carfilzomib, gastrointestinal toxicities remain some of the most common side effects. This review aims to summarize the previous literature on proteasome inhibitor-induced gastrointestinal toxicities, report on recent updates in the field, and investigate possible mechanisms of this toxicity. RECENT FINDINGS Updates in the literature have included a direct comparison of the safety of approved proteasome inhibitors, bortezomib and carfilzomib, reporting less neurotoxicity and similar gastrointestinal toxicity, from carfilzomib when compared with bortezomib. Many recent studies have investigated the safety of orally bioavailable proteasome inhibitors, such as ixazomib and oprozomib. However, little progress has been made in understanding the possible mechanisms of proteasome inhibitor-induced gastrointestinal toxicities. SUMMARY Although recent studies have continued to report gastrointestinal toxicities resulting from proteasome inhibitor treatment, particularly when combined with other agents or when administered orally, the mechanisms of proteasome inhibitor-induced gut toxicity remain largely unexplored. Further studies are needed to investigate the pathophysiology of this toxicity to improve the safety of existing and novel proteasome inhibitors.
Collapse
|
50
|
Gopalakrishnan S, Ediga HH, Reddy SS, Reddy GB, Ismail A. Procyanidin-B2 enriched fraction of cinnamon acts as a proteasome inhibitor and anti-proliferative agent in human prostate cancer cells. IUBMB Life 2018. [DOI: 10.1002/iub.1735] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Srividya Gopalakrishnan
- Department of Endocrinology and Metabolism; National Institute of Nutrition; Hyderabad Telangana India
| | | | - S. Sreenivasa Reddy
- Department of Biochemistry; National Institute of Nutrition; Hyderabad Telangana India
| | - G. Bhanuprakash Reddy
- Department of Biochemistry; National Institute of Nutrition; Hyderabad Telangana India
| | - Ayesha Ismail
- Department of Endocrinology and Metabolism; National Institute of Nutrition; Hyderabad Telangana India
| |
Collapse
|