1
|
Bartolucci D, Pession A, Hrelia P, Tonelli R. Precision Anti-Cancer Medicines by Oligonucleotide Therapeutics in Clinical Research Targeting Undruggable Proteins and Non-Coding RNAs. Pharmaceutics 2022; 14:pharmaceutics14071453. [PMID: 35890348 PMCID: PMC9315662 DOI: 10.3390/pharmaceutics14071453] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/24/2022] [Accepted: 07/08/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer incidence and mortality continue to increase, while the conventional chemotherapeutic drugs confer limited efficacy and relevant toxic side effects. Novel strategies are urgently needed for more effective and safe therapeutics in oncology. However, a large number of proteins are considered undruggable by conventional drugs, such as the small molecules. Moreover, the mRNA itself retains oncological functions, and its targeting offers the double advantage of blocking the tumorigenic activities of the mRNA and the translation into protein. Finally, a large family of non-coding RNAs (ncRNAs) has recently emerged that are also dysregulated in cancer, but they could not be targeted by drugs directed against the proteins. In this context, this review describes how the oligonucleotide therapeutics targeting RNA or DNA sequences, are emerging as a new class of drugs, able to tackle the limitations described above. Numerous clinical trials are evaluating oligonucleotides for tumor treatment, and in the next few years some of them are expected to reach the market. We describe the oligonucleotide therapeutics targeting undruggable proteins (focusing on the most relevant, such as those originating from the MYC and RAS gene families), and for ncRNAs, in particular on those that are under clinical trial evaluation in oncology. We highlight the challenges and solutions for the clinical success of oligonucleotide therapeutics, with particular emphasis on the peculiar challenges that render it arduous to treat tumors, such as heterogeneity and the high mutation rate. In the review are presented these and other advantages offered by the oligonucleotide as an emerging class of biotherapeutics for a new era of precision anti-cancer medicine.
Collapse
Affiliation(s)
| | - Andrea Pession
- Pediatric Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | - Roberto Tonelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
- Correspondence:
| |
Collapse
|
2
|
IL-10 contributes to gemcitabine resistance in extranodal NK/T-cell lymphoma cells via ABCC4. Invest New Drugs 2022; 40:537-545. [DOI: 10.1007/s10637-022-01224-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/14/2022] [Indexed: 11/27/2022]
|
3
|
Wang J, Yang J, Kopeček J. Nanomedicines in B cell-targeting therapies. Acta Biomater 2022; 137:1-19. [PMID: 34687954 PMCID: PMC8678319 DOI: 10.1016/j.actbio.2021.10.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/29/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023]
Abstract
B cells play multiple roles in immune responses related to autoimmune diseases as well as different types of cancers. As such, strategies focused on B cell targeting attracted wide interest and developed intensively. There are several common mechanisms various B cell targeting therapies have relied on, including direct B cell depletion, modulation of B cell antigen receptor (BCR) signaling, targeting B cell survival factors, targeting the B cell and T cell costimulation, and immune checkpoint blockade. Nanocarriers, used as drug delivery vehicles, possess numerous advantages to low molecular weight drugs, reducing drug toxicity, enhancing blood circulation time, as well as augmenting targeting efficacy and improving therapeutic effect. Herein, we review the commonly used targets involved in B cell targeting approaches and the utilization of various nanocarriers as B cell-targeted delivery vehicles. STATEMENT OF SIGNIFICANCE: As B cells are engaged significantly in the development of many kinds of diseases, utilization of nanomedicines in B cell depletion therapies have been rapidly developed. Although numerous studies focused on B cell targeting have already been done, there are still various potential receptors awaiting further investigation. This review summarizes the most relevant studies that utilized nanotechnologies associated with different B cell depletion approaches, providing a useful tool for selection of receptors, agents and/or nanocarriers matching specific diseases. Along with uncovering new targets in the function map of B cells, there will be a growing number of candidates that can benefit from nanoscale drug delivery.
Collapse
Affiliation(s)
- Jiawei Wang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT, United States; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, United States; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.
| |
Collapse
|
4
|
Gupta A, Deka R, Raju S, Singh HB, Butcher RJ. Synthesis of intramolecularly coordinated heteroleptic diorganotellurides and diorganotelluroxides: Isolation of monomeric diorganotelluroxide [{2,6-(Me2NCH2)2C6H3}2TeO] and diorganohydroxytelluronium chloride [{2,6-(Me2NCH2)2C6H3}2Te(OH)]Cl. J Organomet Chem 2019. [DOI: 10.1016/j.jorganchem.2019.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
5
|
Gao L, Han H, Wang H, Cao L, Feng WH. IL-10 knockdown with siRNA enhances the efficacy of Doxorubicin chemotherapy in EBV-positive tumors by inducing lytic cycle via PI3K/p38 MAPK/NF-kB pathway. Cancer Lett 2019; 462:12-22. [PMID: 31352079 DOI: 10.1016/j.canlet.2019.07.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/02/2019] [Accepted: 07/22/2019] [Indexed: 12/19/2022]
Abstract
High levels of IL-10 expression in Epstein-Barr virus (EBV) associated tumors have been reported and it is likely to be important for maintaining EBV latency and EBV-associated tumors. The switch from the latent form of EBV to the lytic form in tumor cells can lead to tumor cell lysis. Here, we found that knockdown of IL-10 induced EBV lytic replication. Subsequently, we demonstrated that IL-10 knockdown activated BZLF1 promoter through PI3K-p38 MAPK-NF-κB signaling pathway. Interestingly, we verified that VEGF-A was required for IL-10 knockdown to activate PI3K signaling and the accompanying EBV lytic induction. Exogenous recombinant human VEGF-A induced PI3K activation and EBV lytic infection, and inhibition of VEGF-A signaling prevented the PI3K/AKT phosphorylation and EBV reactivation responded to IL-10 knockdown. Most importantly, IL-10 knockdown synergized with chemotherapeutic agent Doxorubicin to kill EBV associated tumor cells in vitro and repress EBV-positive tumor growth in vivo. Our results suggest that inhibition of IL-10 has the potential to serve as a new supplemental strategy for the treatment of EBV-associated tumors.
Collapse
Affiliation(s)
- Li Gao
- State Key Laboratory of Agrobiotechnology, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China; China Academy of Medicine Sciences, Peking Union Medical College, Institute of Medicinal Plant Development, Beijing, 100193, China
| | - Haige Han
- State Key Laboratory of Agrobiotechnology, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Honglei Wang
- State Key Laboratory of Agrobiotechnology, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Li Cao
- China Academy of Medicine Sciences, Peking Union Medical College, Institute of Medicinal Plant Development, Beijing, 100193, China
| | - Wen-Hai Feng
- State Key Laboratory of Agrobiotechnology, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
6
|
Limongi T, Susa F, Cauda V. Nanoparticles for hematologic diseases detection and treatment. HEMATOLOGY & MEDICAL ONCOLOGY 2019; 4:1000183. [PMID: 33860108 PMCID: PMC7610588 DOI: 10.15761/hmo.1000183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanotechnology, as an interdisciplinary science, combines engineering, physics, material sciences, and chemistry with the biomedicine knowhow, trying the management of a wide range of diseases. Nanoparticle-based devices holding tumor imaging, targeting and therapy capabilities are formerly under study. Since conventional hematological therapies are sometimes defined by reduced selectivity, low therapeutic efficacy and many side effects, in this review we discuss the potential advantages of the NPs' use in alternative/combined strategies. In the introduction the basic notion of nanomedicine and nanoparticles' classification are described, while in the main text nanodiagnostics, nanotherapeutics and theranostics solutions coming out from the use of a wide-ranging NPs availability are listed and discussed.
Collapse
Affiliation(s)
- Tania Limongi
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Francesca Susa
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Valentina Cauda
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| |
Collapse
|
7
|
Yossipof TE, Bazak ZR, Kenigsbuch-Sredni D, Caspi RR, Kalechman Y, Sredni B. Tellurium Compounds Prevent and Reverse Type-1 Diabetes in NOD Mice by Modulating α4β7 Integrin Activity, IL-1β, and T Regulatory Cells. Front Immunol 2019; 10:979. [PMID: 31191514 PMCID: PMC6549385 DOI: 10.3389/fimmu.2019.00979] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/16/2019] [Indexed: 12/25/2022] Open
Abstract
The study shows that treatment of NOD mice with either of two tellurium-based small molecules, AS101 [ammonium trichloro(dioxoethylene-o,o')tellurate] or SAS [octa-O-bis-(R,R)-tartarate ditellurane] could preserve β cells function and mass. These beneficial effects were reflected in decreased incidence of diabetes, improved glucose clearance, preservation of body weight, and increased survival. The normal glucose levels were associated with increased insulin levels, preservation of β cell mass and increased islet size. Importantly, this protective activity could be demonstrated when the compounds were administered either at the early pre-diabetic phase with no or initial insulitis, at the pre-diabetic stage with advanced insulitis, or even at the advanced, overtly diabetic stage. We further demonstrate that both tellurium compounds prevent migration of autoimmune lymphocytes to the pancreas, via inhibition of the α4β7 integrin activity. Indeed, the decreased migration resulted in diminished pancreatic islets damage both with respect to their size, β cell function, and caspase-3 activity, the hallmark of apoptosis. Most importantly, AS101 and SAS significantly elevated the number of T regulatory cells in the pancreas, thus potentially controlling the autoimmune process. We show that the compounds inhibit pancreatic caspase-1 activity followed by decreased levels of the inflammatory cytokines IL-1β and IL-17 in the pancreas. These properties enable the compounds to increase the proportion of Tregs in the pancreatic lymph nodes. AS101 and SAS have been previously shown to regulate specific integrins through a unique redox mechanism. Our current results suggest that amelioration of disease in NOD mice by this unique mechanism is due to decreased infiltration of pancreatic islets combined with increased immune regulation, leading to decreased inflammation within the islets. As these tellurium compounds show remarkable lack of toxicity in clinical trials (AS101) and pre-clinical studies (SAS), they may be suitable for the treatment of type-1 diabetes.
Collapse
Affiliation(s)
- Tom Eitan Yossipof
- The Mina & Everard Goodman Faculty of Life Sciences, The Safdiè AIDS and Immunology Research Center, C.A.I.R. Institute, Ramat Gan, Israel
| | - Ziva Roy Bazak
- The Mina & Everard Goodman Faculty of Life Sciences, The Safdiè AIDS and Immunology Research Center, C.A.I.R. Institute, Ramat Gan, Israel
| | | | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Yona Kalechman
- The Mina & Everard Goodman Faculty of Life Sciences, The Safdiè AIDS and Immunology Research Center, C.A.I.R. Institute, Ramat Gan, Israel
| | - Benjamin Sredni
- The Mina & Everard Goodman Faculty of Life Sciences, The Safdiè AIDS and Immunology Research Center, C.A.I.R. Institute, Ramat Gan, Israel
| |
Collapse
|
8
|
Chemotherapy and Inflammatory Cytokine Signalling in Cancer Cells and the Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1152:173-215. [PMID: 31456184 DOI: 10.1007/978-3-030-20301-6_9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer is the result of a cell's acquisition of a variety of biological capabilities or 'hallmarks' as outlined by Hanahan and Weinberg. These include sustained proliferative signalling, the ability to evade growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and the ability to invade other tissue and metastasize. More recently, the ability to escape immune destruction has been recognized as another important hallmark of tumours. It is suggested that genome instability and inflammation accelerates the acquisition of a variety of the above hallmarks. Inflammation, is a product of the body's response to tissue damage or pathogen invasion. It is required for tissue repair and host defense, but prolonged inflammation can often be the cause for disease. In a cancer patient, it is often unclear whether inflammation plays a protective or deleterious role in disease progression. Chemotherapy drugs can suppress tumour growth but also induce pathways in tumour cells that have been shown experimentally to support tumour progression or, in other cases, encourage an anti-tumour immune response. Thus, with the goal of better understanding the context under which each of these possible outcomes occurs, recent progress exploring chemotherapy-induced inflammatory cytokine production and the effects of cytokines on drug efficacy in the tumour microenvironment will be reviewed. The implications of chemotherapy on host and tumour cytokine pathways and their effect on the treatment of cancer patients will also be discussed.
Collapse
|
9
|
Reilley MJ, McCoon P, Cook C, Lyne P, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, Fowler N, Curran M, Liu Q, Zhou T, Schmidt J, Jo M, Lee SJ, Yamashita M, Hughes SG, Fayad L, Piha-Paul S, Nadella MVP, Xiao X, Hsu J, Revenko A, Monia BP, MacLeod AR, Hong DS. STAT3 antisense oligonucleotide AZD9150 in a subset of patients with heavily pretreated lymphoma: results of a phase 1b trial. J Immunother Cancer 2018; 6:119. [PMID: 30446007 PMCID: PMC6240242 DOI: 10.1186/s40425-018-0436-5] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/28/2018] [Indexed: 01/05/2023] Open
Abstract
Background The Janus kinase (JAK) and signal transduction and activation of transcription (STAT) signaling pathway is an attractive target in multiple cancers. Activation of the JAK-STAT pathway is important in both tumorigenesis and activation of immune responses. In diffuse large B-cell lymphoma (DLBCL), the transcription factor STAT3 has been associated with aggressive disease phenotype and worse overall survival. While multiple therapies inhibit upstream signaling, there has been limited success in selectively targeting STAT3 in patients. Antisense oligonucleotides (ASOs) represent a compelling therapeutic approach to target difficult to drug proteins such as STAT3 through of mRNA targeting. We report the evaluation of a next generation STAT3 ASO (AZD9150) in a non-Hodgkin’s lymphoma population, primarily consisting of patients with DLBCL. Methods Patients with relapsed or treatment refractory lymphoma were enrolled in this expansion cohort. AZD9150 was administered at 2 mg/kg and the 3 mg/kg (MTD determined by escalation cohort) dose levels with initial loading doses in the first week on days 1, 3, and 5 followed by weekly dosing. Patients were eligible to remain on therapy until unacceptable toxicity or progression. Blood was collected pre- and post-treatment for analysis of peripheral immune cells. Results Thirty patients were enrolled, 10 at 2 mg/kg and 20 at 3 mg/kg dose levels. Twenty-seven patients had DLBCL. AZD9150 was safe and well tolerated at both doses. Common drug-related adverse events included transaminitis, fatigue, and thrombocytopenia. The 3 mg/kg dose level is the recommended phase 2 dose. All responses were seen among DLBCL patients, including 2 complete responses with median duration of response 10.7 months and 2 partial responses. Peripheral blood cell analysis of three patients without a clinical response to therapy revealed a relative increase in proportion of macrophages, CD4+, and CD8+ T cells; this trend did not reach statistical significance. Conclusions AZD9150 was well tolerated and demonstrated efficacy in a subset of heavily pretreated patients with DLBCL. Studies in combination with checkpoint immunotherapies are ongoing. Trial registration Registered at ClinicalTrials.gov: NCT01563302. First submitted 2/13/2012. Electronic supplementary material The online version of this article (10.1186/s40425-018-0436-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Matthew J Reilley
- Division of Hematology/Oncology, University of Virginia Health System, Charlottesville, VA, USA
| | - Patricia McCoon
- Oncology, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Waltham, MA, USA
| | - Carl Cook
- Oncology, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Waltham, MA, USA
| | - Paul Lyne
- Oncology, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Waltham, MA, USA
| | | | - Youngsoo Kim
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Richard Woessner
- Oncology, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Waltham, MA, USA
| | - Anas Younes
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Nathan Fowler
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 0455, Houston, TX, 77030, USA
| | - Michael Curran
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 0455, Houston, TX, 77030, USA
| | - Qinying Liu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 0455, Houston, TX, 77030, USA
| | - Tianyuan Zhou
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Joanna Schmidt
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Minji Jo
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Samantha J Lee
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Mason Yamashita
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Steven G Hughes
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Luis Fayad
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 0455, Houston, TX, 77030, USA
| | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 0455, Houston, TX, 77030, USA
| | - Murali V P Nadella
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Waltham, MA, USA
| | - Xiaokun Xiao
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Jeff Hsu
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Alexey Revenko
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Brett P Monia
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - A Robert MacLeod
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 0455, Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Teixeira ML, Menezes LRA, Barison A, de Oliveira ARM, Piovan L. Investigation of Chemical Stability of Dihalogenated Organotelluranes in Organic-Aqueous Media: The Protagonism of Water. J Org Chem 2018; 83:7341-7346. [PMID: 29373033 DOI: 10.1021/acs.joc.7b02971] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The biological activity of tellurium compounds is closely related to the tellurium oxidation state or some of their structural features. Hypervalent dihalogenated organotelluranes 1-[butyl(dichloro)-λ4-tellanyl]-2-(methoxymethyl)benzene (1a) and 1-[butyl(dibromide)-λ4-tellanyl]-2-(methoxymethyl)benzene (1b) have been described as inhibitors of proteases (cysteine and threonine) and tyrosine phosphatases. However, poor attention has been given to their physicochemical properties. Here, a detailed investigation of the stability in water of these organotelluranes is reported using 125Te NMR analysis. Dihalogenated organotelluranes 1a and 1b were both stable in DMSO- d6 (from 25 to 75 °C), demonstrating their thermal stability. However, the addition of a phosphate buffer solution (pH 2-8) to 1a or 1b resulted in an immediate conversion to a new Te species, assumed to be the corresponding telluroxide. Similar behavior was observed in pure water, demonstrating the low chemical stability of these dihalogenated species in the presence of water. These results allow concluding that previous biological activity reported for dihalogenated organotelluranes 1a and 1b could be attributed to the corresponding derivatives from the reaction with water. In the same way as for AS-101, we demonstrated that organotelluranes 1a and 1b are not stable in aqueous solution. It suggests a proactive role of these organotelluranes in previously reported biological activity.
Collapse
Affiliation(s)
- Mariana L Teixeira
- Department of Chemistry , Universidade Federal do Paraná , Avenida Coronel Francisco Heráclito dos Santos, 100 - Jardim das Américas , Curitiba , Paraná 81531980 , Brazil
| | - Leociley R A Menezes
- Department of Chemistry , Universidade Federal do Paraná , Avenida Coronel Francisco Heráclito dos Santos, 100 - Jardim das Américas , Curitiba , Paraná 81531980 , Brazil
| | - Andersson Barison
- Department of Chemistry , Universidade Federal do Paraná , Avenida Coronel Francisco Heráclito dos Santos, 100 - Jardim das Américas , Curitiba , Paraná 81531980 , Brazil
| | - Alfredo R M de Oliveira
- Department of Chemistry , Universidade Federal do Paraná , Avenida Coronel Francisco Heráclito dos Santos, 100 - Jardim das Américas , Curitiba , Paraná 81531980 , Brazil
| | - Leandro Piovan
- Department of Chemistry , Universidade Federal do Paraná , Avenida Coronel Francisco Heráclito dos Santos, 100 - Jardim das Américas , Curitiba , Paraná 81531980 , Brazil
| |
Collapse
|
11
|
Srivastava K, Panda A, Sharma S, Singh HB. Telluroxanes: Synthesis, structure and applications. J Organomet Chem 2018. [DOI: 10.1016/j.jorganchem.2018.02.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
12
|
Goyal S, Oak E, Luo J, Cashen AF, Carson K, Fehniger T, DiPersio J, Bartlett NL, Wagner-Johnston ND. Minimal activity of nanoparticle albumin-bound (nab) paclitaxel in relapsed or refractory lymphomas: results of a phase-I study. Leuk Lymphoma 2017; 59:357-362. [PMID: 28597723 DOI: 10.1080/10428194.2017.1330954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Compared with solvent-based taxanes, nanoparticle albumin-bound (nab®) paclitaxel has demonstrated improved efficacy and tolerability in several solid tumor malignancies. Studies evaluating nab paclitaxel in patients with lymphoma are lacking. In this planned phase-I/phase-II study, we sought to determine the safety and efficacy of nab-paclitaxel in patients with relapsed/refractory (R/R) lymphoma. Eligible patients (R/R to ≥2 prior systemic therapies) received weekly nab-paclitaxel on days 1, 8 and 15 every 28 days. Dosing was initiated at 100 mg/m2 with dose escalations in 25 mg/m2 increments up to 150 mg/m2 in a classic 3 + 3 design. Twenty heavily pretreated patients (median 5 prior regimens), including 65% with refractory disease, enrolled. The maximum dose tested was well tolerated and grade 3/4 hematologic adverse events (neutropenia 25%, thrombocytopenia 20% and anemia 15%) were modest. The overall response rate was 10% with two partial responses, leading to a decision to close the study prematurely.
Collapse
Affiliation(s)
- Sagun Goyal
- a St. Louis University , St. Louis , MO , USA
| | - Eunhye Oak
- b Siteman Comprehensive Cancer Center, Washington University , St. Louis , MO , USA
| | - Jingqin Luo
- b Siteman Comprehensive Cancer Center, Washington University , St. Louis , MO , USA.,c Division of Public Health Sciences, Department of Surgery , Washington University Biostatistics , St. Louis , MO , USA
| | - Amanda F Cashen
- b Siteman Comprehensive Cancer Center, Washington University , St. Louis , MO , USA
| | - Kenneth Carson
- b Siteman Comprehensive Cancer Center, Washington University , St. Louis , MO , USA
| | - Todd Fehniger
- b Siteman Comprehensive Cancer Center, Washington University , St. Louis , MO , USA
| | - John DiPersio
- b Siteman Comprehensive Cancer Center, Washington University , St. Louis , MO , USA
| | - Nancy L Bartlett
- b Siteman Comprehensive Cancer Center, Washington University , St. Louis , MO , USA
| | | |
Collapse
|
13
|
Hachmo Y, Kalechman Y, Skornick I, Gafter U, Caspi RR, Sredni B. The Small Tellurium Compound AS101 Ameliorates Rat Crescentic Glomerulonephritis: Association with Inhibition of Macrophage Caspase-1 Activity via Very Late Antigen-4 Inactivation. Front Immunol 2017; 8:240. [PMID: 28326083 PMCID: PMC5339302 DOI: 10.3389/fimmu.2017.00240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/20/2017] [Indexed: 11/13/2022] Open
Abstract
Crescentic glomerulonephritis (CGN) is the most aggressive form of GN and, if untreated, patients can progress to end-stage renal failure within weeks of presentation. The α4β1 integrin very late antigen-4 (VLA-4) is an adhesion molecule of fundamental importance to the recruitment of leukocytes in inflammation. We addressed the role of VLA-4 in mediating progressive renal injury in a rat model of CGN using a small tellurium compound. AS101 [ammonium trichloro(dioxoethylene-o,o')tellurate]. This compound has been previously shown to uniquely inhibit VLA-4 activity by redox inactivation of adjacent thiols in the exofacial domain of VLA-4. The study shows that administration of AS101 either before or after glomerular basement membrane anti-serum injection ameliorates crescent formation or preserves renal function. This was associated with profound inhibition of critical inflammatory mediators, accompanied by decreased glomerular infiltration of macrophages. Mechanistic studies demonstrated vla-4 inactivation on glomerular macrophages both in vitro and in vivo as well as inhibition of caspase-1 activity. Importantly, this cysteine protease activity modification was dependent on VLA-4 inactivation and was associated with the anti-inflammatory activity of AS101. We propose that inactivation of macrophage VLA-4 by AS101 in vivo results in a decrease of inflammatory cytokines and chemokines produced in the glomeruli of diseased rats, resulting in decreased further macrophage recruitment and decreased extracellular matrix expansion. Thus, AS101, which is currently in clinical trials for other indications, might be beneficial for treatment of CGN.
Collapse
Affiliation(s)
- Yafit Hachmo
- C.A.I.R. Institute, The Safdiè AIDS and Immunology Research Center, The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan , Israel
| | - Yona Kalechman
- C.A.I.R. Institute, The Safdiè AIDS and Immunology Research Center, The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan , Israel
| | - Itai Skornick
- C.A.I.R. Institute, The Safdiè AIDS and Immunology Research Center, The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan , Israel
| | - Uzi Gafter
- Laboratory of Nephrology and Transplant Immunology, Rabin Medical Center, Petah-Tikva, Israel; Tel Aviv University, Tel Aviv, Israel
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health , Bethesda, MD , USA
| | - Benjamin Sredni
- C.A.I.R. Institute, The Safdiè AIDS and Immunology Research Center, The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan , Israel
| |
Collapse
|
14
|
Shen F, Long D, Yu T, Chen X, Liao Y, Wu Y, Lin X. Vinblastine differs from Taxol as it inhibits the malignant phenotypes of NSCLC cells by increasing the phosphorylation of Op18/stathmin. Oncol Rep 2017; 37:2481-2489. [PMID: 28259950 DOI: 10.3892/or.2017.5469] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/10/2016] [Indexed: 12/20/2022] Open
Abstract
Taxol (paclitaxel) and vinblastine (VBL) are both efficacious chemotherapeutic agents that target the microtubules of tumor cells, but each functions in a mutual antagonistic manner. Op18/stathmin is a small molecular phosphoprotein which promotes depolymerization of microtubules. Non-small cell lung cancer (NSCLC) NCI-H1299 cells were employed to compare the curative effects of VBL and Taxol and explore the correlation between drug sensitivity and Op18/stathmin signaling. The present study found that VBL obviously promoted cellular apoptosis and initiated activation of caspase 3 and 9, and inhibited cell proliferation and colony formation, as well as cell migration in the NCI-H1299 cells in contrast with Taxol. VBL did not affect the expression of Op18/stathmin, but increased its phosphorylation at all 4 serine sites. Conversely, Taxol mainly decreased the expression of Op18/stathmin and the phosphorylation at Ser25 and Ser63 sites. Silencing of Op18/stathmin by RNA interference (RNAi) led to a great reduction in the differences in the cell proliferation inhibition between VBL and Taxol. VBL treatment notably weakened the expression of PP2A, Bcl-2, NF-κB and interleukin-10 (IL-10) and autocrine IL-10 compared with Taxol; whereas PP2A was substantially increased following Taxol induction. High expression of Op18/stathmin was found to be negatively correlated with the sensitivity of Taxol in the NSCLC cells, but had a minor impact on VBL cytotoxicity. These findings revealed that both VBL and Taxol induce cell apoptosis through Op18/stathmin, but the mechanisms are completely different. VBL is an attractive alternative to the treatment of Taxol-resistant tumors with high expression of Op18/stathmin.
Collapse
Affiliation(s)
- Fang Shen
- Department of Medical Laboratory, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Dan Long
- Department of Medical Laboratory, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Ting Yu
- Department of Medical Laboratory, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Xian Chen
- Department of Medical Laboratory, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Ying Liao
- Department of Medical Laboratory, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| | - Yi Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410000, P.R. China
| | - Xuechi Lin
- Department of Medical Laboratory, Changsha Medical University, Changsha, Hunan 410219, P.R. China
| |
Collapse
|
15
|
Wang H, Wang L, Wuxiao Z, Huang H, Jiang W, Li Z, Lu Y, Xia Z. Increased serum levels of interleukin-10 predict poor prognosis in extranodal natural killer/T-cell lymphoma patients receiving asparaginase-based chemotherapy. Onco Targets Ther 2015; 8:2589-99. [PMID: 26396532 PMCID: PMC4576888 DOI: 10.2147/ott.s91077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
There are currently no prognostic biomarkers for extranodal natural killer/T-cell lymphoma (ENKTL) patients receiving asparaginase-based chemotherapy. Interleukin-10 (IL-10) is a pleiotropic cytokine that is involved in the stimulation and suppression of immune responses and influences the prognosis of different subtypes of lymphoma. We retrospectively analyzed 98 newly diagnosed patients with ENKTL receiving asparaginase-based chemotherapy. Baseline serum IL-10 levels were tested with sandwich enzyme-linked immunosorbent assays. Patients with high IL-10 (≥12.28 pg/mL) at diagnosis tended to have more adverse clinical features. Patients with low IL-10 (<12.28 pg/mL) at diagnosis had better progression-free survival (PFS) (P>0.001) and overall survival (OS) (P<0.001). Multivariate analysis revealed that baseline serum IL-10 level ≥12.28 pg/mL, stage III/IV, elevated serum ferritin, and elevated serum Epstein-Barr virus DNA level at diagnosis were four adverse factors for PFS and OS. Based on these four independent prediction factors, we divided the patients into different subgroups as follows: group 1, no adverse factors; group 2, one factor; group 3, two factors; and group 4, three or four factors. Furthermore, significant differences in PFS and OS were found between the groups. Our results suggest that pretreatment serum IL-10 is a novel, powerful predictor of prognosis for ENKTL patients receiving asparaginase-based chemotherapy, which suggests a role for IL-10 in the pathogenesis of this disease and offers new insight into potential therapeutic strategies.
Collapse
Affiliation(s)
- Hua Wang
- Department of Hematological Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China ; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Liang Wang
- Department of Hematological Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China ; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - ZhiJun Wuxiao
- Department of Hematology & Oncology, The Affiliated Hospital, Hainan Medical College, Haikou, Hainan, People's Republic of China
| | - HuiQiang Huang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - WenQi Jiang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - ZhiMing Li
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yue Lu
- Department of Hematological Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China ; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - ZhongJun Xia
- Department of Hematological Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China ; State Key Laboratory of Oncology in South China, Guangzhou, People's Republic of China ; Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| |
Collapse
|