1
|
Joung H, Seo S, Liu H. MG132 induces cell type‑specific anticancer effects in uterine leiomyosarcoma cell lines. Mol Med Rep 2025; 31:159. [PMID: 40211694 PMCID: PMC12015380 DOI: 10.3892/mmr.2025.13524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/14/2025] [Indexed: 04/25/2025] Open
Abstract
Uterine leiomyosarcoma (Ut‑LMS) is a rare and aggressive malignant tumor with limited therapeutic options. Therefore, exploration of novel treatment strategies is necessary. MG132 is a potent proteasome inhibitor that has shown promising potential in cancer therapy by inducing apoptosis through disruption of protein homeostasis. Despite its promising applications in various cancers, its effects on Ut‑LMS remains largely unexplored. Therefore, the present study investigated the effects of MG132 on Ut‑LMS cell lines (SK‑LMS‑1, SK‑UT‑1 and SK‑UT‑1B) in terms of cytotoxicity, apoptosis induction, cell cycle progression, autophagy and reactive oxygen species (ROS) production. Treatment with MG132 (0‑2 µM for 24 h) induced a dose‑dependent reduction in cell viability across all three cell lines, and the lactate dehydrogenase release assays confirmed membrane damage. Moreover, apoptosis induction was assessed using annexin V and 7‑AAD staining, which revealed dose‑dependent apoptosis in all three cell lines. Western blot analysis revealed increased cleaved poly‑adenosine diphosphate ribose polymerase and caspase‑3 levels, thereby indicating activation of the apoptotic pathway in response to MG132 treatment. MG132 also induced G2/M phase arrest in SK‑LMS‑1 and SK‑UT‑1 cells and altered the expression of cell cycle regulatory proteins, such as p21, p27 and p53. Furthermore, MG132 promoted autophagy in all three cell lines by increasing light chain 3 II levels. ROS levels remained unchanged in SK‑LMS‑1 cells but increased in SK‑UT‑1B and SK‑UT‑1 cells. Furthermore, the ROS scavenger N‑acetylcysteine effectively reduced MG132‑induced apoptosis in SK‑UT‑1 cells. These findings highlight the cytotoxicity of MG132 in Ut‑LMS cells, emphasize its potential as a therapeutic agent for Ut‑LMS, provide insights into its mechanisms of action, and suggest possible strategies for improving treatment efficacy.
Collapse
Affiliation(s)
- Hosouk Joung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Hwasun-eup, Jeollanam-do 58128, Republic of Korea
| | - Suho Seo
- Department of Food and Drug, Chosun University Graduate School, Gwangju 61452, Republic of Korea
| | - Hyunju Liu
- Department of Obstetrics and Gynecology, Chosun University College of Medicine, Gwangju 61452, Republic of Korea
- Department of Obstetrics and Gynecology, Chosun University Hospital, Gwangju 61453, Republic of Korea
| |
Collapse
|
2
|
Xiang C, Guo R, Ti J, Zhang S, Wang T. Infection-related adverse events comparison of bortezomib, carfilzomib and ixazomib: a pharmacovigilance study based on FAERS. Expert Opin Drug Saf 2025:1-10. [PMID: 40243068 DOI: 10.1080/14740338.2025.2494692] [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: 10/09/2024] [Revised: 02/21/2025] [Accepted: 03/05/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Bortezomib, carfilzomib and ixazomib are the most common proteasome inhibitors (PIs) used to treat Multiple Myeloma (MM). We conducted a pharmacovigilance analysis using the Food and Drug Administration Adverse Event Reporting System (FAERS), aiming to offer a reference for safe and reasonable clinical use. RESEARCH DESIGN AND METHODS Adverse drug reaction (ADR) signals of bortezomib, carfilzomib and ixazomib were analyzed by statistical methods including Reporting Odds Ratio (ROR), Proportional Reporting Ratios (PRR), and Multi-item Gamma-Poisson Shrinker (MGPS). RESULTS A total of 13,977, 8263 and 13296 ADRs of bortezomib, carfilzomib and ixazomib were analyzed respectively from the FAERS database. The most frequent adverse reaction signal for bortezomib was peripheral neuropathy; for carfilzomib, it was acute kidney injury; for ixazomib, it was vomiting. Then, a total of 43, 23 and 10 infection-related adverse events of bortezomib, carfilzomib and ixazomib were analyzed. The most common infection-related adverse event for bortezomib was cytomegalovirus infection; for carfilzomib, it was bacteremia; and for ixazomib, it was conjunctivitis. CONCLUSIONS In real-world pharmacovigilance studies, PIs are associated with infection-related adverse events, which is crucial for the safe use of PIs in the treatment of MM. However, further research is needed to validate the hypotheses generated in this study.
Collapse
Affiliation(s)
- Chenhuan Xiang
- Department of Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Rongrong Guo
- Department of Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Juanjuan Ti
- Department of Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | | | - Tao Wang
- Department of Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
3
|
Wang Y, Liu YF, Jin SW, Tao Y, Zhang WP, Chen JL, Jiang SF, Mi JQ. Ixazomib or Lenalidomide combined with cyclophosphamide and dexamethasone in the treatment of elderly transplant-ineligible newly diagnosed multiple myeloma. Sci Rep 2025; 15:6538. [PMID: 39994363 PMCID: PMC11850894 DOI: 10.1038/s41598-025-91126-5] [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: 12/30/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
Oral-drug based regimens are useful in certain circumstances for transplant-ineligible newly diagnosed multiple myeloma (TI-NDMM), but few studies have compared Ixazomib based regimen with lenalidomide based regimen head-to-head. We carried out a prospective randomized, open, parallel group trial in patients with TI-NDMM in 3 China centers from March 2020 to December 2022. Sixty-three patients were available for final analysis, ICd (Ixazomib/cyclophosphamide/dexamethasone, n = 31) and RCd (lenalidomide/cyclophosphamide/dexamethasone, n = 32). The primary objective was to compare the two regimens by analyzing the overall response rate (ORR), safety profiles, progression-free survival (PFS) and overall survival (OS). We also explored clinical and the biological characteristics of the patients with primary drug resistance. Baseline characteristics were well balanced between ICd and RCd groups, with the median age 70 vs. 70 years; 12.9% vs. 12.5% of patients had stage III disease; 25.8% vs. 28.1% had high-risk cytogenetic abnormalities. The overall response rate (ORR) at the end of 4 cycles was 87.1% vs. 71.9% (odds ratio [OR], 1.212; 95% CI, 0.938-1.565; P = 0.213); the best ≥ VGPR rate was 41.9% vs. 31.2% (OR, 1.342; 95% CI 0.694-2.597; P = 0.439). Among high-risk cytogenetic patients, ORR was higher in the ICd group, 75% vs. 55.5% (P = 0.620), respectively. After 35 months follow-up, the median PFS were 22 and 23 months between ICd and RCd groups (P = 0.897). Median OS was not reached, estimated 3-year OS rate was 86.4% vs. 85.4% (P = 0.774). The most common adverse events of grade 3 or 4 were neutropenia (6.5% in the ICd group vs. 31.3% in the RCd group), anemia (19.4% vs. 18.8%), pneumonia (0 vs. 15.6%) and diarrhea (12.9% vs. 0). Treatment emergent adverse events (TEAEs) induced dose reduction and discontinuation were 22.6% vs. 37.5% and 3.2% vs. 6.3% in the ICd vs. RCd group, respectively. Exploration data showed that patients with t (4;14) were insensitive to initial RCd treatment. The ICd regimen showed a tendency towards improved ORR compared to RCd regimen. Both ICd and RCd regimens demonstrated less dose reduction and treatment discontinuation, suggesting their tolerability and feasibility for older individuals with TI-NDMM.Trial registration: This study was registered at Chinese Clinical Trial Register (ChiCTR). Trial registration number: ChiCTR2000029863. Date of registration: 15/02/2020.
Collapse
Affiliation(s)
- Yan Wang
- Hematology Department, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai, 200025, China
| | - Yuan-Fang Liu
- Hematology Department, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai, 200025, China
| | - Shi-Wei Jin
- Hematology Department, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai, 200025, China
| | - Yi Tao
- Hematology Department, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai, 200025, China
| | - Wei-Ping Zhang
- Hematology Department, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai, 200025, China
| | - Jian-Lin Chen
- Department of Hematology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China
| | - Song-Fu Jiang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Jian-Qing Mi
- Hematology Department, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, National Research Center for Translational Medicine (Shanghai), State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Shanghai, 200025, China.
| |
Collapse
|
4
|
Luo X, Wu G, Feng J, Zhang J, Fu H, Yu H, Han Z, Nie W, Zhu Z, Liu B, Pan W, Li B, Wang Y, Zhang C, Li T, Zhang W, Wu S. Novel pleuromutilin derivatives conjugated with phenyl-sulfide and boron-containing moieties as potent antibacterial agents against antibiotic-resistant bacteria. Eur J Med Chem 2024; 277:116745. [PMID: 39106659 DOI: 10.1016/j.ejmech.2024.116745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/03/2024] [Accepted: 07/24/2024] [Indexed: 08/09/2024]
Abstract
In response to the escalating threat of microbial resistance, a series of novel pleuromutilin derivatives, conjugated with phenyl-sulfide and boron-containing moieties, were designed and synthesized. Most derivatives, especially 14b and 16b, demonstrated significant efficacy against Gram-positive bacteria, including multidrug-resistant strains, as well as pleuromutilin-resistant strains. Compound 16b showed high stability in the liver microsomes of rats and humans, along with acceptable tolerance in vitro and in vivo. Additionally, compound 16b exhibited promising efficacy in MRSA-infected mouse models. Our data highlight the potential of conjugated pleuromutilin derivatives as valuable agents against drug-resistant bacteria.
Collapse
Affiliation(s)
- Xinyu Luo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Guangxu Wu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
| | - Jing Feng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jie Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hengjian Fu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
| | - Hang Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zunsheng Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wansen Nie
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, 200241, China
| | - Zihao Zhu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Bo Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Weidong Pan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
| | - Beibei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, 200241, China
| | - Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Chi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Tianlei Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Wenxuan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Song Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of New Drug Research and Development, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
5
|
Esposito D, Monti SM, Supuran CT, Winum JY, De Simone G, Alterio V. Exploring the binding mode of phenyl and vinyl boronic acids to human carbonic anhydrases. Int J Biol Macromol 2024; 282:136873. [PMID: 39454912 DOI: 10.1016/j.ijbiomac.2024.136873] [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: 09/03/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Boronic acids are an interesting but still poorly studied class of carbonic anhydrase inhibitors. Previous investigations proved that derivatives incorporating aromatic, arylalkyl, and arylalkenyl moieties are low micromolar to millimolar inhibitors for several α- and β-CAs involved in pathologic states. Here we report a high-resolution X-ray study on two classes of boronic acids (phenyl and vinyl) in complex with hCA II. Our results unambiguously clarify the binding mode of these molecules to the human carbonic anhydrase active site, which occurs through their tetrahedral anionic form, regardless of the nature of the organic scaffold. Data here presented contribute to the understanding of the inhibition mechanism of boronic acids that can be fruitfully used for the rational design of novel and effective isozyme-specific carbonic anhydrase inhibitors.
Collapse
Affiliation(s)
- Davide Esposito
- Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche (IBB-CNR), Via Pietro Castellino, 111, 80131 Naples, Italy
| | - Simona Maria Monti
- Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche (IBB-CNR), Via Pietro Castellino, 111, 80131 Naples, Italy
| | - Claudiu T Supuran
- Department of NEUROFARBA - Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Jean-Yves Winum
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Giuseppina De Simone
- Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche (IBB-CNR), Via Pietro Castellino, 111, 80131 Naples, Italy.
| | - Vincenzo Alterio
- Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche (IBB-CNR), Via Pietro Castellino, 111, 80131 Naples, Italy.
| |
Collapse
|
6
|
Tyrna P, Procyk G, Szeleszczuk Ł, Młynarczuk-Biały I. Different Strategies to Overcome Resistance to Proteasome Inhibitors-A Summary 20 Years after Their Introduction. Int J Mol Sci 2024; 25:8949. [PMID: 39201634 PMCID: PMC11354503 DOI: 10.3390/ijms25168949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Proteasome inhibitors (PIs), bortezomib, carfilzomib, and ixazomib, are the first-line treatment for multiple myeloma (MM). They inhibit cytosolic protein degradation in cells, which leads to the accumulation of misfolded and malfunctioned proteins in the cytosol and endoplasmic reticulum, resulting in cell death. Despite being a breakthrough in MM therapy, malignant cells develop resistance to PIs via different mechanisms. Understanding these mechanisms drives research toward new anticancer agents to overcome PI resistance. In this review, we summarize the mechanism of action of PIs and how MM cells adapt to these drugs to develop resistance. Finally, we explore these mechanisms to present strategies to interfere with PI resistance. The strategies include new inhibitors of the ubiquitin-proteasome system, drug efflux inhibitors, autophagy disruption, targeting stress response mechanisms, affecting survival and cell cycle regulators, bone marrow microenvironment modulation, and immunotherapy. We list potential pharmacological targets examined in in vitro, in vivo, and clinical studies. Some of these strategies have already provided clinicians with new anti-MM medications, such as panobinostat and selinexor. We hope that further exploration of the subject will broaden the range of therapeutic options and improve patient outcomes.
Collapse
Affiliation(s)
- Paweł Tyrna
- Histology and Embryology Students’ Science Association, Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland;
| | - Grzegorz Procyk
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland;
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1 Str., 02-093 Warsaw, Poland;
| | - Izabela Młynarczuk-Biały
- Department of Histology and Embryology, Faculty of Medicine, Warsaw Medical University, Chalubinskiego 5, 02-004 Warsaw, Poland
| |
Collapse
|
7
|
Carullo G, Rossi S, Giudice V, Pezzotta A, Chianese U, Scala P, Carbone S, Fontana A, Panzeca G, Pasquini S, Contri C, Gemma S, Ramunno A, Saponara S, Galvani F, Lodola A, Mor M, Benedetti R, Selleri C, Varani K, Butini S, Altucci L, Vincenzi F, Pistocchi A, Campiani G. Development of Epigenetic Modifiers with Therapeutic Potential in FMS-Related Tyrosine Kinase 3/Internal Tandem Duplication (FLT3/ITD) Acute Myeloid Leukemia and Other Blood Malignancies. ACS Pharmacol Transl Sci 2024; 7:2125-2142. [PMID: 39022363 PMCID: PMC11249625 DOI: 10.1021/acsptsci.4c00208] [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: 04/09/2024] [Revised: 06/04/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024]
Abstract
Blood cancers encompass a group of diseases affecting the blood, bone marrow, or lymphatic system, representing the fourth most commonly diagnosed cancer worldwide. Leukemias are characterized by the dysregulated proliferation of myeloid and lymphoid cells with different rates of progression (acute or chronic). Among the chronic forms, hairy cell leukemia (HCL) is a rare disease, and no drugs have been approved to date. However, acute myeloid leukemia (AML) is one of the most aggressive malignancies, with a low survival rate, especially in cases with FLT3-ITD mutations. Epigenetic modifications have emerged as promising strategies for the treatment of blood cancers. Epigenetic modulators, such as histone deacetylase (HDAC) inhibitors, are increasingly used for targeted cancer therapy. New hydroxamic acid derivatives, preferentially inhibiting HDAC6 (5a-q), were developed and their efficacy was investigated in different blood cancers, including multiple myeloma (MM), HCL, and AML, pointing out their pro-apoptotic effect as the mechanism of cell death. Among the inhibitors described, 5c, 5g, and 5h were able to rescue the hematopoietic phenotype in vivo using the FLT3-ITD zebrafish model of AML. 5c (leuxinostat) proved its efficacy in cells from FLT3-ITD AML patients, promoting marked acetylation of α-tubulin compared to histone H3, thereby confirming HDAC6 as a preferential target for this new class of hydroxamic acid derivatives at the tested doses.
Collapse
Affiliation(s)
- Gabriele Carullo
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Sara Rossi
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Valentina Giudice
- Department
of Medicine, Surgery, Dentistry “Scuola Medica Salernitana”, University of Salerno, Via S. Allende, Baronissi, SA 84081, Italy
| | - Alex Pezzotta
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, LITA, Fratelli Cervi 93, Segrate, MI 20054, Italy
| | - Ugo Chianese
- Department
of Precision Medicine, University of Campania
Luigi Vanvitelli, Via de Crecchio 7, Naples 80138, Italy
| | - Pasqualina Scala
- Department
of Medicine, Surgery, Dentistry “Scuola Medica Salernitana”, University of Salerno, Via S. Allende, Baronissi, SA 84081, Italy
| | - Sabrina Carbone
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, LITA, Fratelli Cervi 93, Segrate, MI 20054, Italy
| | - Anna Fontana
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Giovanna Panzeca
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Silvia Pasquini
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Borsari 46, Ferrara 44121, Italy
| | - Chiara Contri
- Department
of Translational Medicine, University of
Ferrara, Via Borsari 46, Ferrara 44121, Italy
| | - Sandra Gemma
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Anna Ramunno
- Department
of Pharmacy, University of Salerno, Giovanni Paolo II, 132, Fisciano, SA 84084, Italy
| | - Simona Saponara
- Department
of Life Sciences, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Francesca Galvani
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Alessio Lodola
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Marco Mor
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Rosaria Benedetti
- Department
of Precision Medicine, University of Campania
Luigi Vanvitelli, Via de Crecchio 7, Naples 80138, Italy
- Program
of Medical Epigenetics, Vanvitelli Hospital, Naples 80138, Italy
| | - Carmine Selleri
- Department
of Medicine, Surgery, Dentistry “Scuola Medica Salernitana”, University of Salerno, Via S. Allende, Baronissi, SA 84081, Italy
| | - Katia Varani
- Department
of Translational Medicine, University of
Ferrara, Via Borsari 46, Ferrara 44121, Italy
| | - Stefania Butini
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Lucia Altucci
- Department
of Precision Medicine, University of Campania
Luigi Vanvitelli, Via de Crecchio 7, Naples 80138, Italy
- Program
of Medical Epigenetics, Vanvitelli Hospital, Naples 80138, Italy
- Biogem
Institute of Molecular and Genetic Biology, Ariano Irpino 83031, Italy
| | - Fabrizio Vincenzi
- Department
of Translational Medicine, University of
Ferrara, Via Borsari 46, Ferrara 44121, Italy
| | - Anna Pistocchi
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, LITA, Fratelli Cervi 93, Segrate, MI 20054, Italy
| | - Giuseppe Campiani
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
- Bioinformatics
Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-7346, Iran
| |
Collapse
|
8
|
Alrasheed MA, Alamer KA, Albishi M, Alsuhibani AA, Almohammed OA, Alwhaibi A, Almajed AN, Guo JJ. Descriptive Analysis of Adverse Events Reported for New Multiple Myeloma Medications Using FDA Adverse Event Reporting System (FAERS) Databases from 2015 to 2022. Pharmaceuticals (Basel) 2024; 17:815. [PMID: 39065666 PMCID: PMC11279559 DOI: 10.3390/ph17070815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/09/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND New multiple myeloma (MM) medications have revolutionized the treatment landscape, but they are also associated with a range of adverse events (AEs). This study aims to provide a comprehensive overview of AEs reported for four new MM medications: daratumumab, ixazomib, elotuzumab, and panobinostat. METHODS This study uses a descriptive retrospective approach to analyze the FDA Adverse Event Reporting System (FAERS) from 2015 to 2022. It includes variables like medication names, report details, patient demographics, adverse events, and reporter types. The initial dataset consists of over 3700 adverse events, which are categorized into 21 groups for clarity and comparison. RESULTS The FAERS database revealed 367,756 adverse events (AEs) associated with novel multiple myeloma drugs from 2015-2022. Ixazomib had the highest number of reported AEs with 206,243 reports, followed by daratumumab with 98,872 reports, then elotuzumab with 26,193 AEs. Ixazomib's AE reports increased dramatically over the study period, rising approximately 51-fold from 1183 in 2015 to 60,835 in 2022. Of the medications studied, ixazomib also recorded the highest number of deaths (24,206), followed by daratumumab (11,624), panobinostat (7227), and elotuzumab (3349). The majority of AEs occurred in patients aged 55-64 and 65-74 years. CONCLUSIONS Ixazomib, a new MM medication, had the highest number of AEs reported. Also, it has the highest rate of reported deaths compared to other new MM medications. Clinicians should be aware of the potential AEs associated with this medication and further research is needed to understand the reasons for the high number of AEs and to develop mitigation strategies. More attention should also be paid to the safety of new multiple myeloma medications in younger patients.
Collapse
Affiliation(s)
- Marwan A. Alrasheed
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia; (M.A.); (O.A.A.); (A.A.)
| | - Khalid A. Alamer
- Pharmacy Practice Department, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 34221, Saudi Arabia;
| | - Mashael Albishi
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia; (M.A.); (O.A.A.); (A.A.)
| | - Abdulrahman A. Alsuhibani
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Buraidah 51452, Saudi Arabia;
| | - Omar A. Almohammed
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia; (M.A.); (O.A.A.); (A.A.)
| | - Abdulrahman Alwhaibi
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, P.O. Box 2454, Riyadh 11451, Saudi Arabia; (M.A.); (O.A.A.); (A.A.)
| | - Abdullah N. Almajed
- Pharmaceutical Care Division, King Faisal Specialist Hospital and Research Centre, MBC 11, P.O. Box 3354, Riyadh 11211, Saudi Arabia;
| | - Jeff J. Guo
- James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH 45267, USA;
| |
Collapse
|
9
|
Pu J, Liu T, Wang X, Sharma A, Schmidt-Wolf IGH, Jiang L, Hou J. Exploring the role of histone deacetylase and histone deacetylase inhibitors in the context of multiple myeloma: mechanisms, therapeutic implications, and future perspectives. Exp Hematol Oncol 2024; 13:45. [PMID: 38654286 DOI: 10.1186/s40164-024-00507-5] [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: 01/15/2024] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Histone deacetylase inhibitors (HDACis) are a significant category of pharmaceuticals that have developed in the past two decades to treat multiple myeloma. Four drugs in this category have received approval from the U.S. Food and Drug Administration (FDA) for use: Panobinonstat (though canceled by the FDA in 2022), Vorinostat, Belinostat and Romidepsin. The efficacy of this group of drugs is attributed to the disruption of many processes involved in tumor growth through the inhibition of histone deacetylase, and this mode of action leads to significant anti-multiple myeloma (MM) activity. In MM, inhibition of histone deacetylase has many downstream consequences, including suppression of NF-κB signaling and HSP90, upregulation of cell cycle regulators (p21, p53), and downregulation of antiapoptotic proteins including Bcl-2. Furthermore, HDACis have a variety of direct and indirect oxidative effects on cellular DNA. HDAC inhibitors enhance normal immune function, thereby decreasing the proliferation of malignant plasma cells and promoting autophagy. The various biological effects of inhibiting histone deacetylase have a combined or additional impact when used alongside other chemotherapeutic and targeted drugs for multiple myeloma. This helps to decrease resistance to treatment. Combination treatment regimens that include HDACis have become an essential part of the therapy for multiple myeloma. These regimens incorporate drugs from other important classes of anti-myeloma agents, such as immunomodulatory drugs (IMiDs), conventional chemotherapy, monoclonal antibodies, and proteasome inhibitors. This review provides a comprehensive evaluation of the clinical efficacy and safety data pertaining to the currently approved histone deacetylase inhibitors, as well as an explanation of the crucial function of histone deacetylase in multiple myeloma and the characteristics of the different histone deacetylase inhibitors. Moreover, it provides a concise overview of the most recent developments in the use of histone deacetylase inhibitors for treating multiple myeloma, as well as potential future uses in treatment.
Collapse
Affiliation(s)
- Jingjing Pu
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
| | - Ting Liu
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, NRW, Germany
| | - Xuzhen Wang
- Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
| | - Liping Jiang
- Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, Jiangsu, China.
| | - Jian Hou
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
10
|
Meenaghan T, Hayat A, Walpole G, Dowling M. Advances in the management of myeloma: an update. BRITISH JOURNAL OF NURSING (MARK ALLEN PUBLISHING) 2023; 32:S4-S12. [PMID: 37737854 DOI: 10.12968/bjon.2023.32.17.s4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Myeloma is an aggressive B-cell malignancy resulting from an uncontrolled production of plasma cells in the bone marrow. A multitude of drugs and combinations of drugs are now approved for use to treat this complex disease and nurses require knowledge and skills in recognising and managing new side effects associated with these treatments. This article presents an overview of some of the newer and recently approved drugs and the important side effects that have been associated with them. Clinical nurse specialists and advanced nurse practitioners are at the forefront of patients' treatment journeys and play a central role in supporting patients and families to manage side effects. Through this support, patients can continue the treatments for as long as possible with the aim of maintaining a good quality of life.
Collapse
Affiliation(s)
- Teresa Meenaghan
- Advanced Nurse Practitioner, Haematology Department, Galway University Hospital, Ireland
| | - Amjad Hayat
- Consultant Haematologist and Director of Blood and Tissue Establishment and the Stem Cell Lab, Haematology Department, Galway University Hospital, Ireland
| | - Geraldine Walpole
- Advanced Nurse Practitioner, Haematology, Sligo University Hospital, Ireland
| | - Maura Dowling
- Associate Professor, School of Nursing and Midwifery, University of Galway, Ireland
| |
Collapse
|
11
|
Barreto IV, Machado CB, Almeida DB, Pessoa FMCDP, Gadelha RB, Pantoja LDC, Oliveira DDS, Ribeiro RM, Lopes GS, de Moraes Filho MO, de Moraes MEA, Khayat AS, de Oliveira EHC, Moreira-Nunes CA. Kinase Inhibition in Multiple Myeloma: Current Scenario and Clinical Perspectives. Pharmaceutics 2022; 14:pharmaceutics14091784. [PMID: 36145532 PMCID: PMC9506264 DOI: 10.3390/pharmaceutics14091784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a blood cell neoplasm characterized by excessive production of malignant monoclonal plasma cells (activated B lymphocytes) by the bone marrow, which end up synthesizing antibodies or antibody fragments, called M proteins, in excess. The accumulation of this production, both cells themselves and of the immunoglobulins, causes a series of problems for the patient, of a systemic and local nature, such as blood hyperviscosity, renal failure, anemia, bone lesions, and infections due to compromised immunity. MM is the third most common hematological neoplasm, constituting 1% of all cancer cases, and is a disease that is difficult to treat, still being considered an incurable disease. The treatments currently available cannot cure the patient, but only extend their lifespan, and the main and most effective alternative is autologous hematopoietic stem cell transplantation, but not every patient is eligible, often due to age and pre-existing comorbidities. In this context, the search for new therapies that can bring better results to patients is of utmost importance. Protein tyrosine kinases (PTKs) are involved in several biological processes, such as cell growth regulation and proliferation, thus, mutations that affect their functionality can have a great impact on crucial molecular pathways in the cells, leading to tumorigenesis. In the past couple of decades, the use of small-molecule inhibitors, which include tyrosine kinase inhibitors (TKIs), has been a hallmark in the treatment of hematological malignancies, and MM patients may also benefit from TKI-based treatment strategies. In this review, we seek to understand the applicability of TKIs used in MM clinical trials in the last 10 years.
Collapse
Affiliation(s)
- Igor Valentim Barreto
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Caio Bezerra Machado
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | | | - Flávia Melo Cunha de Pinho Pessoa
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Renan Brito Gadelha
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Laudreísa da Costa Pantoja
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil
| | | | | | - Germison Silva Lopes
- Department of Hematology, César Cals General Hospital, Fortaleza 60015-152, CE, Brazil
| | - Manoel Odorico de Moraes Filho
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Maria Elisabete Amaral de Moraes
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - André Salim Khayat
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil
| | - Edivaldo Herculano Correa de Oliveira
- Faculty of Natural Sciences, Institute of Exact and Natural Sciences, Federal University of Pará (UFPA), Rua Augusto Correa, 01, Belém 66075-990, PA, Brazil
- Laboratory of Cytogenomics and Environmental Mutagenesis, Environment Section (SAMAM), Evandro Chagas Institute (IEC), BR 316, KM 7, s/n, Levilândia, Ananindeua 67030-000, PA, Brazil
| | - Caroline Aquino Moreira-Nunes
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil
- Northeast Biotechnology Network (RENORBIO), Itaperi Campus, Ceará State University, Fortaleza 60740-903, CE, Brazil
- Correspondence:
| |
Collapse
|
12
|
Yang C, Zhuang JL. [Analysis of the efficacy and survival of ixazomib-containing regimens in patients with refractory and relapsed multiple myeloma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:680-684. [PMID: 36709154 PMCID: PMC9593011 DOI: 10.3760/cma.j.issn.0253-2727.2022.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Indexed: 01/30/2023]
Affiliation(s)
- C Yang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - J L Zhuang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
13
|
Auner HW, Brown SR, Walker K, Kendall J, Dawkins B, Meads D, Morgan GJ, Kaiser MF, Cook M, Roberts S, Parrish C, Cook G. Ixazomib with cyclophosphamide and dexamethasone in relapsed or refractory myeloma: MUKeight phase II randomised controlled trial results. Blood Cancer J 2022; 12:52. [PMID: 35365598 PMCID: PMC8972903 DOI: 10.1038/s41408-022-00626-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/05/2022] [Accepted: 01/24/2022] [Indexed: 11/15/2022] Open
Abstract
The all-oral combination of ixazomib, cyclophosphamide, and dexamethasone (ICD) is well tolerated and effective in newly diagnosed and relapsed multiple myeloma (MM). We carried out MUKeight, a randomised, controlled, open, parallel group, multi-centre phase II trial in patients with relapsed MM after prior treatment with thalidomide, lenalidomide, and a proteasome inhibitor (ISRCTN58227268), with the primary objective to test whether ICD has improved clinical activity compared to cyclophosphamide and dexamethasone (CD) in terms of progression-free survival (PFS). Between January 2016 and December 2018, 112 participants were randomised between ICD (n = 58) and CD (n = 54) in 33 UK centres. Patients had a median age of 70 years and had received a median of four prior lines of therapy. 74% were classed as frail. Median PFS in the ICD arm was 5.6 months, compared to 6.7 months with CD (hazard ratio (HR) = 1.21, 80% CI 0.9–1.6, p = 0.3634). Response rates and overall survival were not significantly different between ICD and CD. Dose modifications or omissions, and serious adverse events (SAEs), occurred more often in the ICD arm. In summary, the addition of ixazomib to cyclophosphamide and dexamethasone did not improve outcomes in the comparatively frail patients enroled in the MUKeight trial.
Collapse
Affiliation(s)
- Holger W Auner
- Department of Immunology and Inflammation and The Hugh and Josseline Langmuir Centre for Myeloma Research, Imperial College London, London, UK
| | - Sarah R Brown
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Katrina Walker
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Jessica Kendall
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Bryony Dawkins
- Academic Unit of Health Economics, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - David Meads
- Academic Unit of Health Economics, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Gareth J Morgan
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Martin F Kaiser
- The Institute of Cancer Research, London, UK.,The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Mark Cook
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
| | - Sadie Roberts
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Christopher Parrish
- Department of Clinical Haematology, St. James's University Hospital, Leeds, UK
| | - Gordon Cook
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK. .,Department of Clinical Haematology, St. James's University Hospital, Leeds, UK.
| |
Collapse
|
14
|
Šterman A, Sosič I, Časar Z. Primary trifluoroborate-iminiums enable facile access to chiral α-aminoboronic acids via Ru-catalyzed asymmetric hydrogenation and simple hydrolysis of the trifluoroborate moiety. Chem Sci 2022; 13:2946-2953. [PMID: 35432849 PMCID: PMC8905798 DOI: 10.1039/d1sc07065g] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/26/2022] [Indexed: 01/22/2023] Open
Abstract
This work describes the first preparation and application of primary trifluoroborate-iminiums (pTIMs) as a new, easily accessible and valuable class of organoboron derivatives. An array of structurally diverse pTIMs was prepared from potassium acyltrifluoroborates in excellent yields. Highly efficient and enantioselective [(R,R)-TethTsDpen-RuCl] complex-catalyzed hydrogenation of pTIMs provided direct access to chiral primary trifluoroborate-ammoniums (pTAMs). Moreover, facile synthesis of a series of structurally diverse chiral α-aminoboronic acids from chiral pTAMs was accomplished through novel, operationally simple and efficient conversion using hexamethyldisiloxane/aqueous HCl. Using no chromatography at any point, this work allowed easy access to chiral α-aminoboronic acids, as exemplified by the synthesis of optically pure anti-cancer drugs bortezomib and ixazomib.
Collapse
Affiliation(s)
- Andrej Šterman
- Faculty of Pharmacy, University of Ljubljana Aškerčeva cesta 7 SI-1000 Ljubljana Slovenia
| | - Izidor Sosič
- Faculty of Pharmacy, University of Ljubljana Aškerčeva cesta 7 SI-1000 Ljubljana Slovenia
| | - Zdenko Časar
- Faculty of Pharmacy, University of Ljubljana Aškerčeva cesta 7 SI-1000 Ljubljana Slovenia
- Lek Pharmaceuticals d.d., Sandoz Development Center Slovenia Verovškova ulica 57 SI-1526 Ljubljana Slovenia
| |
Collapse
|
15
|
Cao Y, Tu Y, Fu L, Yu Q, Gao L, Zhang M, Zeng L, Zhang C, Shao J, Zhu H, Zhou Y, Li J, Zhang J. Metabolism guided optimization of peptidomimetics as non-covalent proteasome inhibitors for cancer treatment. Eur J Med Chem 2022; 233:114211. [PMID: 35218994 DOI: 10.1016/j.ejmech.2022.114211] [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: 12/06/2021] [Revised: 01/20/2022] [Accepted: 02/17/2022] [Indexed: 11/04/2022]
Abstract
A series of novel non-covalent peptidomimetic proteasome inhibitors possessing bulky group at the C-terminus and N-alkylation at the N-terminus were designed with the aim to increase metabolic stabilities in vivo. All the target compounds were screened for their inhibitory activities against human 20S proteasome, and most analogs exhibited notable potency compared with the positive control bortezomib with IC50 values lower than 10 nM, which also displayed potent cytotoxic activities against multiple myeloma (MM) cell lines and human acute myeloid leukemia (AML) cells. Furthermore, whole blood stability and in vivo proteasome inhibitory activity experiments of selected compounds were conducted for further evaluation, and the representative compound 43 (IC50 = 8.39 ± 2.32 nM, RPMI-8226: IC50 = 15.290 ± 2.281 nM, MM-1S: IC50 = 9.067 ± 3.103 nM, MV-4-11: IC50 = 2.464 ± 0.713 nM) revealed a half-life extension of greater than 9-fold (329.21 min VS 36.79 min) and potent proteasome inhibitory activity in vivo. The positive results confirmed the reliability of the metabolism guided optimization strategy, and the analogs discovered are potential leads for exploring new anti-MM drugs.
Collapse
Affiliation(s)
- Yu Cao
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yutong Tu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liping Fu
- Department of Pharmacy, Shaoxing TCM Hospital Affiliated to Zhejiang Chinese Medical University, Shaoxing, 312000, China
| | - Qian Yu
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lixin Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Pharmaceutical Science, Jiangnan University, Wuxi, 214122, China
| | - Mengmeng Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Linghui Zeng
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, China
| | - Chong Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, China
| | - Jiaan Shao
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, China
| | - Huajian Zhu
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, China
| | - Yubo Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Metria Medica, Chinese Academy of Sciences, Guangdong, 528400, China.
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Metria Medica, Chinese Academy of Sciences, Guangdong, 528400, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310000, China.
| | - Jiankang Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, China.
| |
Collapse
|
16
|
Sak K. Dietary Flavonoids with Catechol Moiety Inhibit Anticancer Action of Bortezomib: What about the other Boronic Acid-based Drugs? Curr Cancer Drug Targets 2022; 22:741-748. [PMID: 35578889 DOI: 10.2174/1568009622666220516102235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/16/2022] [Accepted: 04/04/2022] [Indexed: 11/22/2022]
Abstract
Approval of the first boronic acid group-containing drug, bortezomib, in 2003 for the treatment of multiple myeloma sparked an increased interest of medicinal chemists in boronic acidbased therapeutics. As a result, another boronic acid moiety-harboring medication, ixazomib, was approved in 2015 as a second-generation proteasome inhibitor for multiple myeloma; and dutogliptin is under clinical investigation in combination therapy against myocardial infarction. Moreover, a large number of novel agents with boronic acid elements in their structure are currently in intensive preclinical studies, allowing us to suppose that at least some of them will enter clinical trials in the near future. On the other hand, only some years after bortezomib approval, direct interactions between its boronic acid group and catechol moiety of green tea catechins as well as some other common dietary flavonoids like quercetin and myricetin were discovered, leading to the formation of stable cyclic boronate esters and abolishing the anticancer activities. Although highly relevant, to date, no reports on possible co-effects of catechol group-containing flavonoids with new-generation boronic acidbased drugs can be found. However, this issue cannot be ignored, especially considering the abundance of catechol moiety-harboring flavonoids in both plant-derived food items as well as over-thecounter dietary supplements and herbal products. Therefore, in parallel with the intensified development of boronic acid-based drugs, their possible interactions with catechol groups of plant-derived flavonoids must also be clarified to provide dietary recommendations to patients for maximizing therapeutic benefits. If concurrently consumed flavonoids can indeed antagonize drug efficacy, it may pose a real risk to clinical outcomes.
Collapse
|
17
|
Jayaweera SPE, Wanigasinghe Kanakanamge SP, Rajalingam D, Silva GN. Carfilzomib: A Promising Proteasome Inhibitor for the Treatment of Relapsed and Refractory Multiple Myeloma. Front Oncol 2021; 11:740796. [PMID: 34858819 PMCID: PMC8631731 DOI: 10.3389/fonc.2021.740796] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/21/2021] [Indexed: 01/04/2023] Open
Abstract
The proteasome is crucial for the degradation of intracellular proteins and plays an important role in mediating a number of cell survival and progression events by controlling the levels of key regulatory proteins such as cyclins and caspases in both normal and tumor cells. However, compared to normal cells, cancer cells are more dependent on the ubiquitin proteasome pathway (UPP) due to the accumulation of proteins in response to uncontrolled gene transcription, allowing proteasome to become a potent therapeutic target for human cancers such as multiple myeloma (MM). Up to date, three proteasome inhibitors namely bortezomib (2003), carfilzomib (2012) and ixazomib (2015) have been approved by the US Food and Drug Administration (FDA) for the treatment of patients with relapsed and/or refractory MM. This review mainly focuses on the biochemical properties, mechanism of action, toxicity profile and pivotal clinical trials related to carfilzomib, a second-generation proteasome inhibitor that binds irreversibly with proteasome to overcome the major toxicities and resistance associated with bortezomib.
Collapse
Affiliation(s)
| | | | - Dharshika Rajalingam
- Department of Chemistry, Faculty of Science, University of Colombo, Colombo, Sri Lanka
| | - Gayathri N Silva
- Department of Chemistry, Faculty of Science, University of Colombo, Colombo, Sri Lanka
| |
Collapse
|
18
|
Tripathi BK, Anderman MF, Bhargava D, Boccuzzi L, Qian X, Wang D, Durkin ME, Papageorge AG, de Miguel FJ, Politi K, Walters KJ, Doroshow JH, Lowy DR. Inhibition of cytoplasmic EZH2 induces antitumor activity through stabilization of the DLC1 tumor suppressor protein. Nat Commun 2021; 12:6941. [PMID: 34862367 PMCID: PMC8642553 DOI: 10.1038/s41467-021-26993-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
mRNA expression of the DLC1 tumor suppressor gene is downregulated in many lung cancers and their derived cell lines, with DLC1 protein levels being low or absent. Although the role of increased EZH2 methyltransferase in cancer is usually attributed to its histone methylation, we unexpectedly observed that post-translational destabilization of DLC1 protein is common and attributable to its methylation by cytoplasmic EZH2, leading to CUL-4A ubiquitin-dependent proteasomal degradation of DLC1. Furthermore, siRNA knockdown of KRAS in several lines increases DLC1 protein, associated with a drastic reduction in cytoplasmic EZH2. Pharmacologic inhibition of EZH2, CUL-4A, or the proteasome can increase the steady-state level of DLC1 protein, whose tumor suppressor activity is further increased by AKT and/or SRC kinase inhibitors, which reverse the direct phosphorylation of DLC1 by these kinases. These rational drug combinations induce potent tumor growth inhibition, with markers of apoptosis and senescence, that is highly dependent on DLC1 protein.
Collapse
Affiliation(s)
- Brajendra K Tripathi
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Meghan F Anderman
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Disha Bhargava
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Luciarita Boccuzzi
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Xiaolan Qian
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Dunrui Wang
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Marian E Durkin
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Alex G Papageorge
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Katerina Politi
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Departments of Pathology and Internal Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, CT, USA
| | - Kylie J Walters
- Structural Biophysics Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - James H Doroshow
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Douglas R Lowy
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
19
|
Cummins NW, Baker J, Chakraborty R, Dean PG, Garcia-Rivera E, Krogman A, Kumar S, Kuzmichev YV, Laird GM, Landay A, Lichterfeld M, Mahmood M, Martinson J, Maynes M, Natesampillai S, Rajkumar V, Rassadkina Y, Ritter KD, Rivera CG, Rizza SA, Subramanian K, Tande AJ, Wonderlich ER, Whitaker JA, Zeuli J, Badley AD. Single center, open label dose escalating trial evaluating once weekly oral ixazomib in ART-suppressed, HIV positive adults and effects on HIV reservoir size in vivo. EClinicalMedicine 2021; 42:101225. [PMID: 34901797 PMCID: PMC8639424 DOI: 10.1016/j.eclinm.2021.101225] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Achieving a functional or sterilizing cure for HIV will require identification of therapeutic interventions that reduce HIV reservoir size in infected individuals. Proteasome inhibitors, such as ixazomib, impact multiple aspects of HIV biology including latency, transcription initiation, viral replication, and infected cell killing through the HIV protease - Casp8p41 pathway, resulting in latency reversal and reduced measures of HIV reservoir size ex vivo. METHODS We conducted a phase 1b/2a dose escalating, open label trial of weekly oral ixazomib for 24 weeks in antiretroviral (ART)-suppressed, HIV positive adults (NCT02946047). The study was conducted from March 2017 to August 2019 at two tertiary referral centers in the United States. The primary outcomes were safety and tolerability of oral ixazomib. Secondary outcomes included changes in immunologic markers and estimates of HIV reservoir size after ixazomib treatment. FINDINGS Sixteen participants completed the study. Ixazomib up to 4mg weekly was safe and well-tolerated, yielding no treatment-emergent events above grade 1. In exploratory analyses, ixazomib treatment was associated with detectable viremia that was below the lower limit of quantification (LLQ) in 9 participants, and viremia that was above LLQ in 4 of 16 participants. While treatment was associated with reduced CD4 counts [baseline 783 cells/ mm3 vs. week-24 724 cells/ mm3 p=0.003], there were no changes in markers of cellular activation, exhaustion or inflammation. Total HIV DNA and proviral sequencing were not altered by ixazomib treatment. Intact proviral DNA assay (IPDA) identified intact proviruses in 14 patients pre-treatment, and in 10/14 of those subjects post treatment values were reduced (P=0.068), allowing a calculated intact proviral half life of 0.6 years (95% CI 0.3, 2.5), compared to 7.1 years (95% CI 3.9, 18, p=0.004) in historical controls. Differentiation Quantitative Viral Outgrowth Assays (dQVOA) identified measurable proviruses in 15 subjects pre-treatment; post-treatment values were numerically reduced in 9, but overall differences were not significantly different. INTERPRETATION Our study successfully met its primary endpoint of demonstrating the safety of ixazomib for 24 weeks in HIV infected persons. Exploratory analyses suggest that the effects observed ex vivo of latency reversal and reductions in HIV reservoir size, also occur in vivo. Future controlled studies of ixazomib are warranted. FUNDING This study was funded by Millennium Pharmaceuticals Inc..; the Mayo Clinic Foundation; the National Institutes of Health, including the National Institute of Allergy and Infectious Diseases, Division of AIDS, the National Heart, Lung and Blood Institute, the National Institute of Diabetes and Digestive and Kidney Diseases, the National Institute of Neurological Disorders and Stroke, and the National Institute on Drug Abuse. Mayo Clinic also acknowledges generous funding support from Mr. Joseph T. and Mrs. Michele P. Betten.
Collapse
Affiliation(s)
- Nathan W Cummins
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Jason Baker
- Division of Infectious Diseases, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Rana Chakraborty
- Division of Pediatric Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Patrick G Dean
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ashton Krogman
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Yury V Kuzmichev
- Department of Infectious Disease Research, Southern Research, Frederick, Maryland, USA
| | | | - Alan Landay
- Division of Geriatrics, Rush University Medical Center, Chicago, IL, USA
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT, and Harvard; Brigham and Women's Hospital, Boston, MA, USA
| | - Maryam Mahmood
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey Martinson
- Division of Geriatrics, Rush University Medical Center, Chicago, IL, USA
| | - Mark Maynes
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Yelizaveta Rassadkina
- Ragon Institute of MGH, MIT, and Harvard; Brigham and Women's Hospital, Boston, MA, USA
| | | | | | - Stacey A Rizza
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Krupa Subramanian
- Department of Infectious Disease Research, Southern Research, Frederick, Maryland, USA
| | - Aaron J Tande
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Jennifer A Whitaker
- Division of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - John Zeuli
- Department of Pharmacy, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Corresponding author.
| |
Collapse
|
20
|
Song S, Gao P, Sun L, Kang D, Kongsted J, Poongavanam V, Zhan P, Liu X. Recent developments in the medicinal chemistry of single boron atom-containing compounds. Acta Pharm Sin B 2021; 11:3035-3059. [PMID: 34729302 PMCID: PMC8546671 DOI: 10.1016/j.apsb.2021.01.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/25/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Various boron-containing drugs have been approved for clinical use over the past two decades, and more are currently in clinical trials. The increasing interest in boron-containing compounds is due to their unique binding properties to biological targets; for example, boron substitution can be used to modulate biological activity, pharmacokinetic properties, and drug resistance. In this perspective, we aim to comprehensively review the current status of boron compounds in drug discovery, focusing especially on progress from 2015 to December 2020. We classify these compounds into groups showing anticancer, antibacterial, antiviral, antiparasitic and other activities, and discuss the biological targets associated with each activity, as well as potential future developments.
Collapse
Key Words
- ACTs, artemisinin combination therapies
- ADCs, Acinetobacter-derived cephalosporinases
- AML, acute myeloid leukemia
- AMT, aminopterin
- BLs, β-lactamases
- BNCT, boron neutron capture therapy
- BNNPs, boron nitride nanoparticles
- BNNTs, boron nitride nanotubes
- Boron-containing compounds
- CEs, carboxylesterases
- CIA, collagen-induced arthritis
- COVID-19, coronavirus disease 2019
- ClpP, casein protease P
- Covalent inhibitors
- GSH, glutathione
- HADC1, class I histone deacetylase
- HBV, hepatitis B virus
- HCV, hepatitis C virus
- HIV, human immunodeficiency virus
- LeuRS, leucyl-tRNA synthetase
- Linker components
- MBLs, metal β-lactamases
- MDR-TB, multidrug-resistant tuberculosis
- MERS, Middle East respiratory syndrome
- MIDA, N-methyliminodiacetic acid
- MM, multiple myeloma
- MTX, methotrexate
- Mcl-1, myeloid cell leukemia 1
- Mtb, Mycobacterium tuberculosis
- NA, neuraminidase
- NS5B, non-nucleoside polymerase
- OBORT, oxaborole tRNA capture
- OPs, organophosphate
- PBA, phenylboronic acid
- PDB, Protein Data Bank
- PPI, protein–protein interaction
- Prodrug
- QM, quinone methide
- RA, rheumatoid arthritis
- ROS, reactive oxygen species
- SARS-CoV-2, syndrome coronavirus 2
- SBLs, serine β-lactamases
- SERD, selective estrogen receptor downregulator
- SHA, salicyl hydroxamic acid
- SaClpP, Staphylococcus aureus caseinolytic protease P
- TB, tuberculosis
- TTR, transthyretin
- U4CR, Ugi 4-component reaction
- cUTI, complex urinary tract infection
- dCTPase, dCTPase pyrophosphatase
Collapse
Affiliation(s)
- Shu Song
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan 250012, China
| | - Ping Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan 250012, China
| | - Lin Sun
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan 250012, China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan 250012, China
| | - Jacob Kongsted
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M. DK-5230, Denmark
| | - Vasanthanathan Poongavanam
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M. DK-5230, Denmark
- Corresponding authors. Tel./fax: +86 531 88380270.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan 250012, China
- Corresponding authors. Tel./fax: +86 531 88380270.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan 250012, China
- Corresponding authors. Tel./fax: +86 531 88380270.
| |
Collapse
|
21
|
Sampat PJ, Bisen M, Srivastava N, Rao S, Gentile T. Accidental Ixazomib Overdose in a Patient With Multiple Myeloma. J Investig Med High Impact Case Rep 2021; 9:23247096211013230. [PMID: 33980053 PMCID: PMC8127745 DOI: 10.1177/23247096211013230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Multiple myeloma is the second most common hematological malignancy. Ixazomib is the first oral proteasome inhibitor approved in the United States for the management of multiple myeloma who have received at least one prior treatment. The availability of oral chemotherapeutic agents for the management of multiple myeloma has made it easier for patients who do not have to come to the hospital for chemotherapy infusions. However, many barriers are associated with oral chemotherapy, and one of them is a misinterpretation of instruction which can have deleterious effects. In this case report, we present a case of a 69-year-old male with multiple myeloma who accidentally took ixazomib daily for 3 days instead of the weekly regimen and thus coming into the hospital with an overdose. In this report, we focus on the adverse effects associated with ixazomib toxicity and how to manage the adverse reactions. Although there is no antidote available for ixazomib, supportive care is very essential in these patients.
Collapse
Affiliation(s)
| | | | | | - Suman Rao
- SUNY Upstate Medical University, Syracuse, NY, USA
| | | |
Collapse
|
22
|
Guo M, Ding P, Zhu Z, Fan L, Zhou Y, Yang S, Yang Y, Gu C. Targeting RFWD2 as an Effective Strategy to Inhibit Cellular Proliferation and Overcome Drug Resistance to Proteasome Inhibitor in Multiple Myeloma. Front Cell Dev Biol 2021; 9:675939. [PMID: 33968945 PMCID: PMC8097052 DOI: 10.3389/fcell.2021.675939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/30/2021] [Indexed: 12/15/2022] Open
Abstract
The potential to overcome resistance to proteasome inhibitors is greatly related with ubiquitin-proteasome system during multiple myeloma (MM) treatment process. The constitutive photomorphogenic 1 (RFWD2), referred to an E3 ubiquitin ligase, has been identified as an oncogene in multiple cancers, yet important questions on the role of RFWD2 in MM biology and treatment remain unclear. Here we demonstrated that MM patients with elevated RFWD2 expression achieved adverse outcome and drug resistance by analyzing gene expression profiling. Moreover, we proved that RFWD2 participated in the process of cell cycle, cell growth and death in MM by mass spectrometry analysis. In vitro study indicated that inducible knockdown of RFWD2 hindered cellular growth and triggered apoptosis in MM cells. Mechanism study revealed that RFWD2 controlled MM cellular proliferation via regulating the degradation of P27 rather than P53. Further exploration unveiled that RFWD2 meditated P27 ubiquitination via interacting with RCHY1, which served as an E3 ubiquitin ligase of P27. Finally, in vivo study illustrated that blocking RFWD2 in BTZ-resistant MM cells overcame the drug resistance in a myeloma xenograft mouse model. Taken together, these findings provide compelling evidence for prompting that targeting RFWD2 may be an effective strategy to inhibit cellular proliferation and overcome drug resistance to proteasome inhibitor in MM.
Collapse
Affiliation(s)
- Mengjie Guo
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Large Data Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Pinggang Ding
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhen Zhu
- College of Health and Rehabilitation & College of Acupuncture and Massage, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lu Fan
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanyan Zhou
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shu Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ye Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunyan Gu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Large Data Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
23
|
Liang X, Wu P, Yang Q, Xie Y, He C, Yin L, Yin Z, Yue G, Zou Y, Li L, Song X, Lv C, Zhang W, Jing B. An update of new small-molecule anticancer drugs approved from 2015 to 2020. Eur J Med Chem 2021; 220:113473. [PMID: 33906047 DOI: 10.1016/j.ejmech.2021.113473] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/02/2021] [Accepted: 04/09/2021] [Indexed: 01/09/2023]
Abstract
A high incidence of cancer has given rise to the development of more anti-tumor drugs. From 2015 to 2020, fifty-six new small-molecule anticancer drugs, divided into ten categories according to their anti-tumor target activities, have been approved. These include TKIs (30 drugs), MAPK inhibitors (3 drugs), CDK inhibitors (3 drugs), PARP inhibitors (3 drugs), PI3K inhibitors (3 drugs), SMO receptor antagonists (2 drugs), AR antagonists (2 drugs), SSTR inhibitors (2 drugs), IDH inhibitors (2 drugs) and others (6 drugs). Among them, PTK inhibitors (30/56) have led to a paradigm shift in cancer treatment with less toxicity and more potency. Each of their structures, approval statuses, applications, SAR analyses, and original research synthesis routes have been summarized, giving us a more comprehensive map for further efforts to design more specific targeted agents for reducing cancer in the future. We believe this review will help further research of potential antitumor agents in clinical usage.
Collapse
Affiliation(s)
- Xiaoxia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China.
| | - Pan Wu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Qian Yang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Yunyu Xie
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Changliang He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Lizi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Guizhou Yue
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Cheng Lv
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Wei Zhang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| | - Bo Jing
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, PR China
| |
Collapse
|
24
|
Dunphy K, Dowling P, Bazou D, O’Gorman P. Current Methods of Post-Translational Modification Analysis and Their Applications in Blood Cancers. Cancers (Basel) 2021; 13:1930. [PMID: 33923680 PMCID: PMC8072572 DOI: 10.3390/cancers13081930] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/04/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Post-translational modifications (PTMs) add a layer of complexity to the proteome through the addition of biochemical moieties to specific residues of proteins, altering their structure, function and/or localization. Mass spectrometry (MS)-based techniques are at the forefront of PTM analysis due to their ability to detect large numbers of modified proteins with a high level of sensitivity and specificity. The low stoichiometry of modified peptides means fractionation and enrichment techniques are often performed prior to MS to improve detection yields. Immuno-based techniques remain popular, with improvements in the quality of commercially available modification-specific antibodies facilitating the detection of modified proteins with high affinity. PTM-focused studies on blood cancers have provided information on altered cellular processes, including cell signaling, apoptosis and transcriptional regulation, that contribute to the malignant phenotype. Furthermore, the mechanism of action of many blood cancer therapies, such as kinase inhibitors, involves inhibiting or modulating protein modifications. Continued optimization of protocols and techniques for PTM analysis in blood cancer will undoubtedly lead to novel insights into mechanisms of malignant transformation, proliferation, and survival, in addition to the identification of novel biomarkers and therapeutic targets. This review discusses techniques used for PTM analysis and their applications in blood cancer research.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Paul Dowling
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland;
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland;
| |
Collapse
|
25
|
Ixazomib as treatment for pyoderma gangrenosum associated with IgA smoldering multiple myeloma. Ann Hematol 2021; 101:441-442. [PMID: 33860355 DOI: 10.1007/s00277-021-04524-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
|
26
|
Up-front ixazomib in older patients with myeloma. Blood 2021; 137:3584-3586. [PMID: 34196682 DOI: 10.1182/blood.2021011262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
27
|
Roeten MS, van Meerloo J, Kwidama ZJ, ter Huizen G, Segerink WH, Zweegman S, Kaspers GJ, Jansen G, Cloos J. Pre-Clinical Evaluation of the Proteasome Inhibitor Ixazomib against Bortezomib-Resistant Leukemia Cells and Primary Acute Leukemia Cells. Cells 2021; 10:665. [PMID: 33802801 PMCID: PMC8002577 DOI: 10.3390/cells10030665] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/19/2021] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
At present, 20-30% of children with acute leukemia still relapse from current chemotherapy protocols, underscoring the unmet need for new treatment options, such as proteasome inhibition. Ixazomib (IXA) is an orally available proteasome inhibitor, with an improved safety profile compared to Bortezomib (BTZ). The mechanism of action (proteasome subunit inhibition, apoptosis induction) and growth inhibitory potential of IXA vs. BTZ were tested in vitro in human (BTZ-resistant) leukemia cell lines. Ex vivo activity of IXA vs. BTZ was analyzed in 15 acute lymphoblastic leukemia (ALL) and 9 acute myeloid leukemia (AML) primary pediatric patient samples. BTZ demonstrated more potent inhibitory effects on constitutive β5 and immunoproteasome β5i proteasome subunit activity; however, IXA more potently inhibited β1i subunit than BTZ (70% vs. 29% at 2.5 nM). In ALL/AML cell lines, IXA conveyed 50% growth inhibition at low nanomolar concentrations, but was ~10-fold less potent than BTZ. BTZ-resistant cells (150-160 fold) displayed similar (100-fold) cross-resistance to IXA. Finally, IXA and BTZ exhibited anti-leukemic effects for primary ex vivo ALL and AML cells; mean LC50 (nM) for IXA: 24 ± 11 and 30 ± 8, respectively, and mean LC50 for BTZ: 4.5 ± 1 and 11 ± 4, respectively. IXA has overlapping mechanisms of action with BTZ and showed anti-leukemic activity in primary leukemic cells, encouraging further pre-clinical in vivo evaluation.
Collapse
Affiliation(s)
- Margot S.F. Roeten
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Johan van Meerloo
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Zinia J. Kwidama
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Giovanna ter Huizen
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Wouter H. Segerink
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Sonja Zweegman
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| | - Gertjan J.L. Kaspers
- Princess Maxima Center of Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
- Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology, 1105 AZ Amsterdam, The Netherlands
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands;
| | - Jacqueline Cloos
- Cancer Center Amsterdam, Department of Hematology, Amsterdam UMC, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands; (M.S.F.R.); (J.v.M.); (Z.J.K.); (G.t.H.); (W.H.S.); (S.Z.)
| |
Collapse
|
28
|
Soysouvanh F, Giuliano S, Habel N, El-Hachem N, Pisibon C, Bertolotto C, Ballotti R. An Update on the Role of Ubiquitination in Melanoma Development and Therapies. J Clin Med 2021; 10:jcm10051133. [PMID: 33800394 PMCID: PMC7962844 DOI: 10.3390/jcm10051133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/27/2022] Open
Abstract
The ubiquitination system plays a critical role in regulation of large array of biological processes and its alteration has been involved in the pathogenesis of cancers, among them cutaneous melanoma, which is responsible for the most deaths from skin cancers. Over the last decades, targeted therapies and immunotherapies became the standard therapeutic strategies for advanced melanomas. However, despite these breakthroughs, the prognosis of metastatic melanoma patients remains unoptimistic, mainly due to intrinsic or acquired resistances. Many avenues of research have been investigated to find new therapeutic targets for improving patient outcomes. Because of the pleiotropic functions of ubiquitination, and because each step of ubiquitination is amenable to pharmacological targeting, much attention has been paid to the role of this process in melanoma development and resistance to therapies. In this review, we summarize the latest data on ubiquitination and discuss the possible impacts on melanoma treatments.
Collapse
Affiliation(s)
- Frédéric Soysouvanh
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
| | - Serena Giuliano
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
| | - Nadia Habel
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
| | - Najla El-Hachem
- Laboratory of Cancer Signaling, University of Liège, 4020 Liège, Belgium;
| | - Céline Pisibon
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
| | - Corine Bertolotto
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
- Equipe labellisée Fondation ARC 2019, 06200 Nice, France
| | - Robert Ballotti
- Inserm U1065, C3M, Team 1, Biology, and Pathologies of Melanocytes, University of Nice Côte d’Azur, 06200 Nice, France; (F.S.); (S.G.); (N.H.); (C.P.); (C.B.)
- Equipe labellisée Ligue Contre le Cancer 2020, 06200 Nice, France
- Correspondence: ; Tel.: +33-4-89-06-43-32
| |
Collapse
|
29
|
Wang Q, Dong Z, Su J, Huang J, Xiao P, Tian L, Chen Y, Ma L, Chen X. Ixazomib inhibits myeloma cell proliferation by targeting UBE2K. Biochem Biophys Res Commun 2021; 549:1-7. [PMID: 33647537 DOI: 10.1016/j.bbrc.2021.02.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/11/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE Ixazomib is a selective, effective, and reversible inhibitor of 20S proteasome and is approved for the treatment of multiple myeloma. Ubiquitin-conjugating enzyme E2 (UBE2K) is involved in the synthesis of K48-linked ubiquitin chains and is the target of certain drugs used for the treatment of tumors. The purpose of this study was to investigate the relationship between ixazomib and UBE2K in myeloma cells. METHODS We used CCK-8 and Annexin V-FITC/propidium iodide kit to detect the effects of ixazomib on survival and apoptosis of RPMI-8226 and U-266 myeloma cell lines. Quantitative polymerase chain reaction and western blot were used to detect the change in gene and protein expression levels of myeloma cells treated with ixazomib. Furthermore, the regulatory effects of ixazomib on UBE2K and its downstream targets were investigated following the overexpression of UBE2K. RESULTS In myeloma cells, ixazomib decreased cell survival and increased apoptosis in a dose-dependent manner. Ixazomib significantly increased the expression of HIST1H2BD, MNAT1, NEK3, and TARS2, while decreasing the expression of HSPA1B and UBE2K. In addition, ixazomib inhibited the proliferation of myeloma cells, blocked cell cycle, induced cell apoptosis, and increased the production of reactive oxygen species by inhibiting UBE2K expression. Lastly, ixazomib regulates mitosis- and apoptosis-related genes by lowering UBE2K expression. CONCLUSION In summary, ixazomib leads to impaired proliferation of myeloma cells by targeting UBE2K.
Collapse
Affiliation(s)
- Qingqing Wang
- Blood Rheumatism Immunology Department, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China.
| | - Zhigao Dong
- Blood Rheumatism Immunology Department, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China.
| | - Junnan Su
- Blood Rheumatism Immunology Department, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China.
| | - Jinmei Huang
- Blood Rheumatism Immunology Department, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China.
| | - Pingping Xiao
- Blood Rheumatism Immunology Department, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China.
| | - Lihong Tian
- Blood Rheumatism Immunology Department, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China.
| | - Yongquan Chen
- Blood Rheumatism Immunology Department, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China.
| | - Lili Ma
- Blood Rheumatism Immunology Department, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China.
| | - Xuyan Chen
- Blood Rheumatism Immunology Department, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian, China.
| |
Collapse
|
30
|
Lemoine J, Bonnin A, Marjanovic Z, van de Wyngaert Z, Ikhlef S, Alsuliman T, M'Hammedi-Bouzina F, Mohty M, Malard F. Resolution of bortezomib-associated chalazia/blepharitis after switch to ixazomib: A case report. Curr Res Transl Med 2021; 69:103283. [PMID: 33639586 DOI: 10.1016/j.retram.2021.103283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/08/2021] [Indexed: 01/12/2023]
Affiliation(s)
- Jean Lemoine
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Agnes Bonnin
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Zora Marjanovic
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Zoe van de Wyngaert
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Souhila Ikhlef
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Tamim Alsuliman
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Fella M'Hammedi-Bouzina
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Mohamad Mohty
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Florent Malard
- Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, Sorbonne Université, INSERM UMRs 938, Paris, France.
| |
Collapse
|
31
|
Liu JR, Li J, Huang XJ. [Problems and progress of autologous hematopoietic stem cell transplantation in multiple myeloma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:82-86. [PMID: 33677876 PMCID: PMC7957257 DOI: 10.3760/cma.j.issn.0253-2727.2021.01.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Indexed: 12/22/2022]
Affiliation(s)
- J R Liu
- Department of Hematology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - J Li
- Department of Hematology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - X J Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| |
Collapse
|
32
|
Gu Y, Barwick BG, Shanmugam M, Hofmeister CC, Kaufman J, Nooka A, Gupta V, Dhodapkar M, Boise LH, Lonial S. Downregulation of PA28α induces proteasome remodeling and results in resistance to proteasome inhibitors in multiple myeloma. Blood Cancer J 2020; 10:125. [PMID: 33318477 PMCID: PMC7736847 DOI: 10.1038/s41408-020-00393-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/14/2020] [Accepted: 10/28/2020] [Indexed: 01/05/2023] Open
Abstract
Protein homeostasis is critical for maintaining eukaryotic cell function as well as responses to intrinsic and extrinsic stress. The proteasome is a major portion of the proteolytic machinery in mammalian cells and plays an important role in protein homeostasis. Multiple myeloma (MM) is a plasma cell malignancy with high production of immunoglobulins and is especially sensitive to treatments that impact protein catabolism. Therapeutic agents such as proteasome inhibitors have demonstrated significant benefit for myeloma patients in all treatment phases. Here, we demonstrate that the 11S proteasome activator PA28α is upregulated in MM cells and is key for myeloma cell growth and proliferation. PA28α also regulates MM cell sensitivity to proteasome inhibitors. Downregulation of PA28α inhibits both proteasomal load and activity, resulting in a change in protein homeostasis less dependent on the proteasome and leads to cell resistance to proteasome inhibitors. Thus, our findings suggest an important role of PA28α in MM biology, and also provides a new approach for targeting the ubiquitin-proteasome system and ultimately sensitivity to proteasome inhibitors.
Collapse
Affiliation(s)
- Yanyan Gu
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Benjamin G Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Craig C Hofmeister
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Jonathan Kaufman
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Ajay Nooka
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Vikas Gupta
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Madhav Dhodapkar
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Road, Atlanta, GA, 30322, USA. .,Winship Cancer Institute, Emory University, 1365 Clifton Road, Atlanta, GA, 30322, USA.
| |
Collapse
|
33
|
Bai Y, Su X. Updates to the drug-resistant mechanism of proteasome inhibitors in multiple myeloma. Asia Pac J Clin Oncol 2020; 17:29-35. [PMID: 32920949 DOI: 10.1111/ajco.13459] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022]
Abstract
Proteasome inhibitors (PIs) have been a kind of backbone therapies for newly diagnosed as well as relapsed or refractory myeloma patients in the last two decades. Bortezomib, the first-in-class PI, was approved by the United States Food and Drug Administration in 2003. The key roles of this class of agents are targeting at the overstressed 26S proteasome, which are involved in the pathogenesis of the disease. Despite recent advancements in clinical antimyeloma treatment, the acquisition of resistance is a major limitation in PI therapy. This review aims at a better understanding of the pathways and biomarkers involved in MM drug resistance.
Collapse
Affiliation(s)
- Yang Bai
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Xing Su
- The Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| |
Collapse
|
34
|
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: 67] [Impact Index Per Article: 13.4] [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
|
35
|
Dash AB, Zhang J, Shen L, Li B, Berg D, Lin J, Avet‐Loiseau H, Bahlis NJ, Moreau P, Richardson PG, Di Bacco A. Clinical benefit of ixazomib plus lenalidomide-dexamethasone in myeloma patients with non-canonical NF-κB pathway activation. Eur J Haematol 2020; 105:274-285. [PMID: 32350909 PMCID: PMC7497066 DOI: 10.1111/ejh.13435] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/23/2020] [Accepted: 04/27/2020] [Indexed: 01/27/2023]
Abstract
OBJECTIVES Evaluating potential relationships between progression-free survival (PFS) and tumor gene expression patterns and mutational status was an exploratory objective of the phase 3 TOURMALINE-MM1 study (NCT01564537) of ixazomib-lenalidomide-dexamethasone (IRd) vs placebo-Rd in 722 patients with relapsed/refractory multiple myeloma (MM). METHODS We utilized gene expression and mutation data from screening bone marrow aspirates to identify tumors with non-canonical nuclear factor-κB (NF-κB) signaling pathway activation. RESULTS DNA/RNA sequencing data were available for 339 (47.0%)/399 (55.2%) patients; 49/339 (14.5%) patients had non-canonical NF-κB pathway gene mutations (tumor-necrosis-factor receptor-associated factor 2, 3 [TRAF2, TRAF3], baculoviral-inhibitor-of-apoptosis repeat-containing 2/3 [BIRC2/3]), and PFS was significantly longer with IRd vs placebo-Rd in these patients (hazard ratio [HR] 0.23). In patients with lower TRAF3 expression (median not reached vs 11 months, HR 0.47) and higher NF-κB-inducing kinase (NIK) expression (median not reached vs 14 months, HR 0.45), both associated with non-canonical NF-κB pathway activation, PFS was significantly longer with IRd vs placebo-Rd. TRAF3 expression was decreased in patients harboring t(4;14) and 1q21 amplification, suggesting increased non-canonical NF-κB pathway activation. CONCLUSIONS Adding ixazomib to Rd provides clinical benefit in MM tumors with increased non-canonical NF-κB pathway activity. This is a potential mechanism for activity in 1q21 amplified high-risk tumors.
Collapse
Affiliation(s)
- Ajeeta B. Dash
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | - Jacob Zhang
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | - Lei Shen
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | - Bin Li
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | - Deborah Berg
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | - Jianchang Lin
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| | | | | | - Philippe Moreau
- Department of HematologyUniversity Hospital Hôtel DieuUniversity of NantesNantesFrance
| | | | - Alessandra Di Bacco
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company LimitedCambridgeMAUSA
| |
Collapse
|
36
|
Yang J, Li D, Zhou J. Histone Deacetylase 6 as a Therapeutic Target in B cell-associated Hematological Malignancies. Front Pharmacol 2020; 11:971. [PMID: 32676030 PMCID: PMC7333221 DOI: 10.3389/fphar.2020.00971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
B lymphocytes play a critical role in humoral immunity. Abnormal B cell development and function cause a variety of hematological malignancies such as myeloma, B cell lymphoma, and leukemia. Histone deacetylase 6 (HDAC6) inhibitors alone or in combination with other drugs have shown efficacy in several hematological malignancies, including those resistant to targeted therapies. Mechanistically, HDAC6 inhibitors promote malignant tumor cell apoptosis by inhibiting protein degradation, reinvigorating anti-tumor immunity, and inhibiting cell survival signaling pathways. Due to their specificity, HDAC6 inhibitors represent a very promising and feasible new development pipeline for high-efficacy drugs with limited side effects. This article reviews recent progress in the mechanisms of action of HDAC6 inhibitors for the treatment of B cell-associated hematological malignancies, such as multiple myeloma and B cell non-Hodgkin lymphoma, which are often resistant to targeted therapies.
Collapse
Affiliation(s)
- Jia Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.,Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
37
|
The E3 ligase HUWE1 inhibition as a therapeutic strategy to target MYC in multiple myeloma. Oncogene 2020; 39:5001-5014. [PMID: 32523091 PMCID: PMC7329634 DOI: 10.1038/s41388-020-1345-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/19/2022]
Abstract
Proteasome inhibitors have provided a significant advance in the treatment of multiple myeloma (MM). Consequently, there is increasing interest in developing strategies to target E3 ligases, de-ubiquitinases, and/or ubiquitin receptors within the ubiquitin proteasome pathway, with an aim to achieve more specificity and reduced side-effects. Previous studies have shown a role for the E3 ligase HUWE1 in modulating c-MYC, an oncogene frequently dysregulated in MM. Here we investigated HUWE1 in MM. We identified elevated expression of HUWE1 in MM compared with normal cells. Small molecule-mediated inhibition of HUWE1 resulted in growth arrest of MM cell lines without significantly effecting the growth of normal bone marrow cells, suggesting a favorable therapeutic index. Studies using a HUWE1 knockdown model showed similar growth inhibition. HUWE1 expression positively correlated with MYC expression in MM bone marrow cells and correspondingly, genetic knockdown and biochemical inhibition of HUWE1 reduced MYC expression in MM cell lines. Proteomic identification of HUWE1 substrates revealed a strong association of HUWE1 with metabolic processes in MM cells. Intracellular glutamine levels are decreased in the absence of HUWE1 and may contribute to MYC degradation. Finally, HUWE1 depletion in combination with lenalidomide resulted in synergistic anti-MM activity in both in vitro and in vivo models. Taken together, our data demonstrate an important role of HUWE1 in MM cell growth and provides preclinical rationale for therapeutic strategies targeting HUWE1 in MM.
Collapse
|
38
|
Sánchez G, Chalmers S, Ahumada X, Montecinos L, Olmedo I, Eisner V, Riveros A, Kogan MJ, Lavandero S, Pedrozo Z, Donoso P. Inhibition of chymotrypsin-like activity of the proteasome by ixazomib prevents mitochondrial dysfunction during myocardial ischemia. PLoS One 2020; 15:e0233591. [PMID: 32453773 PMCID: PMC7250417 DOI: 10.1371/journal.pone.0233591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 05/10/2020] [Indexed: 01/02/2023] Open
Abstract
The heart is critically dependent on mitochondrial respiration for energy supply. Ischemia decreases oxygen availability, with catastrophic consequences for cellular energy systems. After a few minutes of ischemia, the mitochondrial respiratory chain halts, ATP levels drop and ion gradients across cell membranes collapse. Activation of cellular proteases and generation of reactive oxygen species by mitochondria during ischemia alter mitochondrial membrane permeability, causing mitochondrial swelling and fragmentation and eventually cell death. The mitochondria, therefore, are important targets of cardioprotection against ischemic injury. We have previously shown that ixazomib (IXA), a proteasome inhibitor used for treating multiple myeloma, effectively reduced the size of the infarct produced by global ischemia in isolated rat hearts and prevented degradation of the sarcoplasmic reticulum calcium release channel RyR2. The aim of this work was to further characterize the protective effect of IXA by determining its effect on mitochondrial morphology and function after ischemia. We also quantified the effect of IXA on levels of mitofusin-2, a protein involved in maintaining mitochondrial morphology and mitochondria-SR communication. We found that mitochondria were significantly preserved and functional parameters such as oxygen consumption, the ability to generate a membrane potential, and glutathione content were improved in mitochondria isolated from hearts perfused with IXA prior to ischemia. IXA also blocked the release of cytochrome c observed in ischemia and significantly preserved mitofusin-2 integrity. These beneficial effects resulted in a significant decrease in the left ventricular end diastolic pressure upon reperfusion and a smaller infarct in isolated hearts.
Collapse
Affiliation(s)
- Gina Sánchez
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- * E-mail: (GS); (PD)
| | - Stefanie Chalmers
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Xavier Ahumada
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Luis Montecinos
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ivonne Olmedo
- Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Veronica Eisner
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ana Riveros
- Departamento de Química Farmacológica y Toxicológica, Facultad Ciencias Químicas y Farmacéuticas Universidad de Chile, Santiago, Chile
| | - Marcelo J. Kogan
- Departamento de Química Farmacológica y Toxicológica, Facultad Ciencias Químicas y Farmacéuticas Universidad de Chile, Santiago, Chile
| | - Sergio Lavandero
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Bioquímica y Biología Molecular, Facultad Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| | - Zully Pedrozo
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| | - Paulina Donoso
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- * E-mail: (GS); (PD)
| |
Collapse
|
39
|
Harris MA, Miles MA, Shekhar TM, Cerra C, Georgy SR, Ryan SD, Cannon CM, Hawkins CJ. The Proteasome Inhibitor Ixazomib Inhibits the Formation and Growth of Pulmonary and Abdominal Osteosarcoma Metastases in Mice. Cancers (Basel) 2020; 12:cancers12051207. [PMID: 32403415 PMCID: PMC7281181 DOI: 10.3390/cancers12051207] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022] Open
Abstract
Osteosarcoma is the most common form of primary bone cancer. Over 20% of osteosarcoma patients present with pulmonary metastases at diagnosis, and nearly 70% of these patients fail to respond to treatment. Previous work revealed that human and canine osteosarcoma cell lines are extremely sensitive to the therapeutic proteasome inhibitor bortezomib in vitro. However, bortezomib has proven disappointingly ineffective against solid tumors including sarcomas in animal experiments and clinical trials. Poor tumor penetration has been speculated to account for the inconsistency between in vitro and in vivo responses of solid tumors to bortezomib. Here we show that the second-generation proteasome inhibitor ixazomib, which reportedly has enhanced solid tumor penetration compared to bortezomib, is toxic to human and canine osteosarcoma cells in vitro. We used experimental osteosarcoma metastasis models to compare the efficacies of ixazomib and bortezomib against primary tumors and metastases derived from luciferase-expressing KRIB or 143B human osteosarcoma cell lines in athymic mice. Neither proteasome inhibitor reduced the growth of primary intramuscular KRIB tumors, however both drugs inhibited the growth of established pulmonary metastases created via intravenous inoculation with KRIB cells, which were significantly better vascularized than the primary tumors. Only ixazomib slowed metastases from KRIB primary tumors and inhibited the growth of 143B pulmonary and abdominal metastases, significantly enhancing the survival of mice intravenously injected with 143B cells. Taken together, these results suggest ixazomib exerts better single agent activity against osteosarcoma metastases than bortezomib. These data provide hope that incorporation of ixazomib, or other proteasome inhibitors that penetrate efficiently into solid tumors, into current regimens may improve outcomes for patients diagnosed with metastatic osteosarcoma.
Collapse
Affiliation(s)
- Michael A. Harris
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Victoria, Australia; (M.A.H.); (M.A.M.); (T.M.S.); (C.C.)
| | - Mark A. Miles
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Victoria, Australia; (M.A.H.); (M.A.M.); (T.M.S.); (C.C.)
| | - Tanmay M. Shekhar
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Victoria, Australia; (M.A.H.); (M.A.M.); (T.M.S.); (C.C.)
| | - Carmelo Cerra
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Victoria, Australia; (M.A.H.); (M.A.M.); (T.M.S.); (C.C.)
| | - Smitha R. Georgy
- Department of Anatomic Pathology, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, 3010 Victoria, Australia;
| | - Stewart D. Ryan
- Translational Research and Animal Clinical Trial Study Group (TRACTS), Faculty of Veterinary and Agricultural Sciences, University of Melbourne, 3010 Melbourne, Australia; (S.D.R.); (C.M.C.)
| | - Claire M. Cannon
- Translational Research and Animal Clinical Trial Study Group (TRACTS), Faculty of Veterinary and Agricultural Sciences, University of Melbourne, 3010 Melbourne, Australia; (S.D.R.); (C.M.C.)
| | - Christine J. Hawkins
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, 3086 Victoria, Australia; (M.A.H.); (M.A.M.); (T.M.S.); (C.C.)
- Correspondence: ; Tel.: +61-3-9479-2339
| |
Collapse
|
40
|
Lai KP, Chen J, Tse WKF. Role of Deubiquitinases in Human Cancers: Potential Targeted Therapy. Int J Mol Sci 2020; 21:ijms21072548. [PMID: 32268558 PMCID: PMC7177317 DOI: 10.3390/ijms21072548] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Deubiquitinases (DUBs) are involved in various cellular functions. They deconjugate ubiquitin (UBQ) from ubiquitylated substrates to regulate their activity and stability. Studies on the roles of deubiquitylation have been conducted in various cancers to identify the carcinogenic roles of DUBs. In this review, we evaluate the biological roles of DUBs in cancer, including proliferation, cell cycle control, apoptosis, the DNA damage response, tumor suppression, oncogenesis, and metastasis. This review mainly focuses on the regulation of different downstream effectors and pathways via biochemical regulation and posttranslational modifications. We summarize the relationship between DUBs and human cancers and discuss the potential of DUBs as therapeutic targets for cancer treatment. This review also provides basic knowledge of DUBs in the development of cancers and highlights the importance of DUBs in cancer biology.
Collapse
Affiliation(s)
- Keng Po Lai
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541004, China;
| | - Jian Chen
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541004, China;
- Correspondence: (J.C.); (W.K.F.T.); Tel.: +86-773-5895810 (J.C.); +81-92-802-4767 (W.K.F.T.)
| | - William Ka Fai Tse
- Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
- Correspondence: (J.C.); (W.K.F.T.); Tel.: +86-773-5895810 (J.C.); +81-92-802-4767 (W.K.F.T.)
| |
Collapse
|
41
|
New generation drugs for treatment of multiple myeloma. Drug Discov Today 2019; 25:367-379. [PMID: 31765717 DOI: 10.1016/j.drudis.2019.11.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 11/08/2019] [Accepted: 11/15/2019] [Indexed: 12/28/2022]
Abstract
Multiple myeloma (MM), a plasma cell malignancy, is characterised by lesions in multiple bones involving transformed, matured post-follicular B cells. The course of the disease involves an initial development of monoclonal gammopathy of undetermined significance (MGUS), followed by smouldering MM, before the full MM disease emerges. Despite novel therapies, MM remains incurable, managed by combination therapies, including proteasome inhibitors (PIs), immunomodulators (IMiDs) and anti-human CD38 (daratumumab). MM patients have an increased risk of thromboembolic events due to combination treatments with IMiDs, PIs and anti-human CD38 antibody, and steroids. This review will examine the efficacy and pro-thrombotic effects of MM therapies.
Collapse
|
42
|
Nicolaou KC, Rigol S. The Role of Organic Synthesis in the Emergence and Development of Antibody–Drug Conjugates as Targeted Cancer Therapies. Angew Chem Int Ed Engl 2019; 58:11206-11241. [DOI: 10.1002/anie.201903498] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Indexed: 12/14/2022]
Affiliation(s)
- K. C. Nicolaou
- Department of ChemistryBioScience Research CollaborativeRice University 6100 Main Street Houston Texas 77005 USA
| | - Stephan Rigol
- Department of ChemistryBioScience Research CollaborativeRice University 6100 Main Street Houston Texas 77005 USA
| |
Collapse
|
43
|
Nicolaou KC, Rigol S. Die Bedeutung der organischen Synthese bei der Entstehung und Entwicklung von Antikörper‐Wirkstoff‐Konjugaten als gezielte Krebstherapien. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201903498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- K. C. Nicolaou
- Department of ChemistryBioScience Research CollaborativeRice University 6100 Main Street Houston TX 77005 USA
| | - Stephan Rigol
- Department of ChemistryBioScience Research CollaborativeRice University 6100 Main Street Houston TX 77005 USA
| |
Collapse
|
44
|
Song Y, Park PMC, Wu L, Ray A, Picaud S, Li D, Wimalasena VK, Du T, Filippakopoulos P, Anderson KC, Qi J, Chauhan D. Development and preclinical validation of a novel covalent ubiquitin receptor Rpn13 degrader in multiple myeloma. Leukemia 2019; 33:2685-2694. [PMID: 30962579 DOI: 10.1038/s41375-019-0467-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 03/25/2019] [Indexed: 12/13/2022]
Abstract
Proteasome inhibition is an effective treatment for multiple myeloma (MM); however, targeting different components of the ubiquitin-proteasome system (UPS) remains elusive. Our RNA-interference studies identified proteasome-associated ubiquitin-receptor Rpn13 as a mediator of MM cell growth and survival. Here, we developed the first degrader of Rpn13, WL40, using a small-molecule-induced targeted protein degradation strategy to selectively degrade this component of the UPS. WL40 was synthesized by linking the Rpn13 covalent inhibitor RA190 with the cereblon (CRBN) binding ligand thalidomide. We show that WL40 binds to both Rpn13 and CRBN and triggers degradation of cellular Rpn13, and is therefore first-in-class in exploiting a covalent inhibitor for the development of degraders. Biochemical and cellular studies show that WL40-induced Rpn13 degradation is both CRBN E3 ligase- and Rpn13-dependent. Importantly, WL40 decreases viability in MM cell lines and patient MM cells, even those resistant to bortezomib. Mechanistically, WL40 interrupts Rpn13 function and activates caspase apoptotic cascade, ER stress response and p53/p21 signaling. In animal model studies, WL40 inhibits xenografted human MM cell growth and prolongs survival. Overall, our data show the development of the first UbR Rpn13 degrader with potent anti-MM activity, and provide proof of principle for the development of degraders targeting components of the UPS for therapeutic application.
Collapse
Affiliation(s)
- Yan Song
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Paul M C Park
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Lei Wu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Arghya Ray
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Deyao Li
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Virangika K Wimalasena
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Ting Du
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Dharminder Chauhan
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|