1
|
Bi J, Zeng J, Liu X, Mo C, Yao M, Zhang J, Yuan P, Jia B, Xu S. Drug delivery for age-related bone diseases: From therapeutic targets to common and emerging therapeutic strategies. Saudi Pharm J 2024; 32:102209. [PMID: 39697472 PMCID: PMC11653637 DOI: 10.1016/j.jsps.2024.102209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
With the accumulation of knowledge on aging, people have gradually realized that among the many factors that cause individual aging, the accumulation of aging cells is an essential cause of organ degeneration and, ultimately, age-related diseases. Most cells present in the bone microenvironment gradually age over time, leading to an imbalance of osteogenesis, osteoclastogenesis, adipogenesis, and chondrogenesis. This imbalance contributes to age-related bone loss and the development of age-related bone diseases, such as osteoporosis. Bone aging can prolong the lifespan and delay the development of age-related diseases. Nanoparticles have controllable and stable physical and chemical properties and can precisely target different tissues and organs. By preparing multiple easily modified and biocompatible nanoparticles as different drug delivery carriers, specifically targeting various diseased tissues for controlled-release and sustained-release administration, the delivery efficiency of drugs can be significantly improved, and the toxicity and side effects of drugs can be substantially reduced, thereby improving the therapeutic effect of age-related bone diseases. In addition, other novel anti-aging strategies (such as stem cell exosomes) also have significant scientific and practical significance in anti-aging research on age-related bone diseases. This article reviews the research progress of various nano-drug-loaded particles and emerging anti-aging methods for treating age-related bone diseases, offering new insights and directions for precise targeted clinical therapies.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohao Liu
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingyan Yao
- Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Jing Zhang
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Wang T, Cui S, Lyu C, Wang Z, Li Z, Han C, Liu W, Wang Y, Xu R. Molecular precision medicine: Multi-omics-based stratification model for acute myeloid leukemia. Heliyon 2024; 10:e36155. [PMID: 39263156 PMCID: PMC11388765 DOI: 10.1016/j.heliyon.2024.e36155] [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: 02/25/2024] [Revised: 08/01/2024] [Accepted: 08/11/2024] [Indexed: 09/13/2024] Open
Abstract
Acute myeloid leukemia (AML), as the most common malignancy of the hematopoietic system, poses challenges in treatment efficacy, relapse, and drug resistance. In this study, we have utilized 151 RNA sequencing datasets, 194 DNA methylation datasets, and 200 somatic mutation datasets from the AML cohort in the TCGA database to develop a multi-omics stratification model. This model enables comparison of prognosis, clinical features, gene mutations, immune microenvironment and drug sensitivity across subgroups. External validation datasets have been sourced from the GEO database, which includes 562 mRNA datasets and 136 miRNA datasets from 984 adult AML patients. Through multi-omics-based stratification model, we classified 126 AML patients into 4 clusters (CS). CS4 had the best prognosis, with the youngest age, highest M3 subtype proportion, fewest copy number alterations, and common mutations in WT1, FLT3, and KIT genes. It showed sensitivity to HDAC inhibitors and BCL-2 inhibitors. Both the M3 subtype and CS4 were identified as independent protective factors for survival. Conversely, CS3 had the worst prognosis due to older age, high copy number alterations, and frequent mutations in RUNX1, DNMT3A, and TP53 genes. Additionally, it showed higher proportions of cytotoxic cells and Tregs, suggesting potential sensitivity to mTOR inhibitors. CS1 had a better prognosis than CS2, with more copy number alterations, while CS2 had higher monocyte proportions. CS1 showed good sensitivity to cytarabine, while CS2 was sensitive to RXR agonists. Both CS1 and CS2, which predominantly featured mutations in FLT3, NPM1, and DNMT3A genes, benefited from FLT3 inhibitors. Using the Kappa test, our stratification model underwent robust validation in the miRNA and mRNA external validation datasets. With advancements in sequencing technology and machine learning algorithms, AML is poised to transition towards multi-omics precision medicine in the future. We aspire for our study to offer new perspectives on multi-drug combination clinical trials and multi-targeted precision medicine for AML.
Collapse
Affiliation(s)
- Teng Wang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Siyuan Cui
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Shandong, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
| | - Chunyi Lyu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenzhen Wang
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Shandong, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
| | - Zonghong Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chen Han
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Weilin Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Wang
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Shandong, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
| | - Ruirong Xu
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Shandong, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, 250014, China
| |
Collapse
|
3
|
Eiken AP, Schmitz E, Drengler EM, Smith AL, Skupa SA, Mohan K, Rana S, Singh S, Mallareddy JR, Mathew G, Natarajan A, El-Gamal D. The Novel Anti-Cancer Agent, SpiD3, Is Cytotoxic in CLL Cells Resistant to Ibrutinib or Venetoclax. HEMATO 2024; 5:321-339. [PMID: 39450301 PMCID: PMC11500768 DOI: 10.3390/hemato5030024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Background B-cell receptor (BCR) signaling is a central driver in chronic lymphocytic leukemia (CLL), along with the activation of pro-survival pathways (e.g., NF-κB) and aberrant anti-apoptotic mechanisms (e.g., BCL2) culminating to CLL cell survival and drug resistance. Front-line targeted therapies such as ibrutinib (BTK inhibitor) and venetoclax (BCL2 inhibitor) have radically improved CLL management. Yet, persisting CLL cells lead to relapse in ~20% of patients, signifying the unmet need of inhibitor-resistant refractory CLL. SpiD3 is a novel spirocyclic dimer of analog 19 that displays NF-κB inhibitory activity and preclinical anti-cancer properties. Recently, we have shown that SpiD3 inhibits CLL cell proliferation and induces cytotoxicity by promoting futile activation of the unfolded protein response (UPR) pathway and generation of reactive oxygen species (ROS), resulting in the inhibition of protein synthesis in CLL cells. Methods We performed RNA-sequencing using CLL cells rendered resistant to ibrutinib and venetoclax to explore potential vulnerabilities in inhibitor-resistant and SpiD3-treated CLL cells. Results The transcriptomic analysis of ibrutinib- or venetoclax-resistant CLL cell lines revealed ferroptosis, UPR signaling, and oxidative stress to be among the top pathways modulated by SpiD3 treatment. By examining SpiD3-induced protein aggregation, ROS production, and ferroptosis in inhibitor-resistant CLL cells, our findings demonstrate cytotoxicity following SpiD3 treatment in cell lines resistant to current front-line CLL therapeutics. Conclusions Our results substantiate the development of SpiD3 as a novel therapeutic agent for relapsed/refractory CLL disease.
Collapse
Affiliation(s)
- Alexandria P. Eiken
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Elizabeth Schmitz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Erin M. Drengler
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Audrey L. Smith
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sydney A. Skupa
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kabhilan Mohan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sandeep Rana
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sarbjit Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jayapal Reddy Mallareddy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Grinu Mathew
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dalia El-Gamal
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
4
|
Liu L, Mo W, Chen M, Qu Y, Wang P, Liang Y, Yan X. Targeted inhibition of DHODH is synergistic with BCL2 blockade in HGBCL with concurrent MYC and BCL2 rearrangement. BMC Cancer 2024; 24:761. [PMID: 38918775 PMCID: PMC11197201 DOI: 10.1186/s12885-024-12534-w] [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: 10/27/2023] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
High-grade B-cell lymphoma (HGBCL), the subtype of non-Hodgkin lymphoma, to be relapsed or refractory in patients after initial therapy or salvage chemotherapy. Dual dysregulation of MYC and BCL2 is one of the important pathogenic mechanisms. Thus, combined targeting of MYC and BCL2 appears to be a promising strategy. Dihydroorotate dehydrogenase (DHODH) is the fourth rate-limiting enzyme for the de novo biosynthesis of pyrimidine. It has been shown to be a potential therapeutic target for multiple diseases. In this study, the DHODH inhibitor brequinar exhibited growth inhibition, cell cycle blockade, and apoptosis promotion in HGBCL cell lines with MYC and BCL2 rearrangements. The combination of brequinar and BCL2 inhibitors venetoclax had a synergistic inhibitory effect on the survival of DHL cells through different pathways. Venetoclax could upregulate MCL-1 and MYC expression, which has been reported as a resistance mechanism of BCL2 inhibitors. Brequinar downregulated MCL-1 and MYC, which could potentially overcome drug resistance to venetoclax in HGBCL cells. Furthermore, brequinar could downregulate a broad range of genes, including ribosome biosynthesis genes, which might contribute to its anti-tumor effects. In vivo studies demonstrated synergetic tumor growth inhibition in xenograft models with brequinar and venetoclax combination treatment. These results provide preliminary evidence for the rational combination of DHODH and BCL2 blockade in HGBCL with abnormal MYC and BCL2.
Collapse
Affiliation(s)
- Lin Liu
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Wenbin Mo
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Miao Chen
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yi Qu
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Pingping Wang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Ying Liang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaojing Yan
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
- , No. 155, North Nanjing Road, Heping District, Shenyang, 110001, China.
| |
Collapse
|
5
|
Yang H, Li D, Gao G. Kaempferol Alleviates Hepatic Injury in Nonalcoholic Steatohepatitis (NASH) by Suppressing Neutrophil-Mediated NLRP3-ASC/TMS1-Caspase 3 Signaling. Molecules 2024; 29:2630. [PMID: 38893506 PMCID: PMC11173805 DOI: 10.3390/molecules29112630] [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: 03/31/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a significant hepatic condition that has gained worldwide attention. Kaempferol (Kae), renowned for its diverse biological activities, including anti-inflammatory, antioxidant, anti-aging, and cardio-protective properties, has emerged as a potential therapeutic candidate for non-alcoholic steatohepatitis (NASH). Despite its promising therapeutic potential, the precise underlying mechanism of Kae's beneficial effects in NASH remains unclear. Therefore, this study aims to clarify the mechanism by conducting comprehensive in vivo and in vitro experiments. RESULTS In this study, a murine model of non-alcoholic steatohepatitis (NASH) was established by feeding C57BL/6 female mice a high-fat diet for 12 weeks. Kaempferol (Kae) was investigated for its ability to modulate systemic inflammatory responses and lipid metabolism in this model (20 mg/kg per day). Notably, Kae significantly reduced the expression of NLRP3-ASC/TMS1-Caspase 3, a crucial mediator of liver tissue inflammation. Additionally, in a HepG2 cell model induced with palmitic acid/oleic acid (PA/OA) to mimic NASH conditions, Kae demonstrated the capacity to decrease lipid droplet accumulation and downregulate the expression of NLRP3-ASC/TMS1-Caspase 3 (20 µM and the final concentration to 20 nM). These findings suggest that Kae may hold therapeutic potential in the treatment of NASH by targeting inflammatory and metabolic pathways. CONCLUSIONS These findings suggest that kaempferol holds potential as a promising therapeutic intervention for ameliorating non-alcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- He Yang
- Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | | | - Guolan Gao
- Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
6
|
Chatzikalil E, Roka K, Diamantopoulos PT, Rigatou E, Avgerinou G, Kattamis A, Solomou EE. Venetoclax Combination Treatment of Acute Myeloid Leukemia in Adolescents and Young Adult Patients. J Clin Med 2024; 13:2046. [PMID: 38610812 PMCID: PMC11012941 DOI: 10.3390/jcm13072046] [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: 02/24/2024] [Revised: 03/25/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Over the past two decades, the prognosis in adolescents and young adults (AYAs) diagnosed with acute myeloid leukemia (AML) has significantly improved. The standard intensive cytotoxic treatment approach for AYAs with AML, consisting of induction chemotherapy with anthracycline/cytarabine combination followed by consolidation chemotherapy or stem cell transplantation, has lately been shifting toward novel targeted therapies, mostly in the fields of clinical trials. One of the most recent advances in treating AML is the combination of the B-cell lymphoma 2 (Bcl-2) inhibitor venetoclax with hypomethylating agents, which has been studied in elderly populations and was approved by the Food and Drug Administration (FDA) for patients over 75 years of age or patients excluded from intensive chemotherapy induction schemas due to comorbidities. Regarding the AYA population, venetoclax combination therapy could be a therapeutic option for patients with refractory/relapsed (R/R) AML, although data from real-world studies are currently limited. Venetoclax is frequently used by AYAs diagnosed with advanced hematologic malignancies, mainly acute lymphoblastic leukemia and myelodysplastic syndromes, as a salvage therapeutic option with considerable efficacy and safety. Herein, we aim to summarize the evidence obtained from clinical trials and observational studies on venetoclax use in AYAs with AML. Based on the available evidence, venetoclax is a safe and effective therapeutic option for R/R AML AYA patients. However, further research in larger cohorts is needed to confirm these data, establishing the benefits of a venetoclax-based regimen for this special population.
Collapse
Affiliation(s)
- Elena Chatzikalil
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (E.C.); (K.R.); (E.R.); (G.A.); (A.K.)
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Kleoniki Roka
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (E.C.); (K.R.); (E.R.); (G.A.); (A.K.)
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Panagiotis T. Diamantopoulos
- First Department of Internal Medicine, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece;
| | - Efthymia Rigatou
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (E.C.); (K.R.); (E.R.); (G.A.); (A.K.)
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Georgia Avgerinou
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (E.C.); (K.R.); (E.R.); (G.A.); (A.K.)
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Antonis Kattamis
- Division of Pediatric Hematology-Oncology, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (E.C.); (K.R.); (E.R.); (G.A.); (A.K.)
- “Aghia Sofia” Children’s Hospital ERN-PeadCan Center, 11527 Athens, Greece
| | - Elena E. Solomou
- Department of Internal Medicine, University of Patras Medical School, 26500 Rion, Greece
| |
Collapse
|
7
|
Chen Q, Wang Y, Shi C, Tong M, Sun H, Dong M, Liu S, Wang L. Molecular Mechanism of the Asarum-Angelica Drug Pair in the Treatment of Periodontitis Based on Network Pharmacology and Experimental Verification. Int J Mol Sci 2023; 24:17389. [PMID: 38139216 PMCID: PMC10744231 DOI: 10.3390/ijms242417389] [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: 10/31/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
(1) To examine the potential mechanism of the Asarum-Angelica drug pair against periodontitis and provide an experimental basis for the treatment of periodontitis with herbal medicine. (2) The core components and core targets of the Asarum-Angelica drug pair in the treatment of periodontitis were detected according to network pharmacology methods. Finally, the effect of the Asarum-Angelica drug pair on osteogenic differentiation was observed in mouse embryonic osteoblast precursor cells. (3) According to the results of network pharmacology, there are 10 potential active ingredients in the Asarum-Angelica drug pair, and 44 potential targets were obtained by mapping the targets with periodontitis treatment. Ten potential active ingredients, such as kaempferol and β-sitosterol, may play a role in treating periodontitis. Cell experiments showed that the Asarum-Angelica drug pair can effectively promote the expression of osteoblast markers alkaline phosphatase (ALP), Runt-related Transcription Factor 2 (RUNX2), and BCL2 mRNA and protein in an inflammatory environment (p < 0.05). (4) Network pharmacology effectively analyzed the molecular mechanism of Asarum-Angelica in the treatment of periodontitis, and the Asarum-Angelica drug pair can promote the differentiation of osteoblasts.
Collapse
Affiliation(s)
- Qianyang Chen
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian 116044, China
| | - Yuhan Wang
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian 116044, China
| | - Chun Shi
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian 116044, China
| | - Meichen Tong
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian 116044, China
| | - Haibo Sun
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian 116044, China
| | - Ming Dong
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian 116044, China
| | - Shuo Liu
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian 116044, China
| | - Lina Wang
- Department of Endodontics and Periodontics, College of Stomatology, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian 116044, China
- Academician Laboratory of Immune and Oral Development and Regeneration, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
8
|
Olejarz W, Basak G. Emerging Therapeutic Targets and Drug Resistance Mechanisms in Immunotherapy of Hematological Malignancies. Cancers (Basel) 2023; 15:5765. [PMID: 38136311 PMCID: PMC10741639 DOI: 10.3390/cancers15245765] [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: 10/31/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
CAR-T cell therapy has revolutionized the treatment of hematological malignancies with high remission rates in the case of ALL and NHL. This therapy has some limitations such as long manufacturing periods, persistent restricted cell sources and high costs. Moreover, combination regimens increase the risk of immune-related adverse events, so the identification new therapeutic targets is important to minimize the risk of toxicities and to guide more effective approaches. Cancer cells employ several mechanisms to evade immunosurveillance, which causes resistance to immunotherapy; therefore, a very important therapeutic approach is to focus on the development of rational combinations of targeted therapies with non-overlapping toxicities. Recent progress in the development of new inhibitory clusters of differentiation (CDs), signaling pathway molecules, checkpoint inhibitors, and immunosuppressive cell subsets and factors in the tumor microenvironment (TME) has significantly improved anticancer responses. Novel strategies regarding combination immunotherapies with CAR-T cells are the most promising approach to cure cancer.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland;
| |
Collapse
|
9
|
Xie J, Wang H, Huang Q, Lin J, Wen H, Miao Y, Lv L, Ruan D, Yu X, Qin L, Zhou Y. Enhanced cytotoxicity to lung cancer cells by mitochondrial delivery of camptothecin. Eur J Pharm Sci 2023; 189:106561. [PMID: 37562549 DOI: 10.1016/j.ejps.2023.106561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/12/2023] [Accepted: 08/06/2023] [Indexed: 08/12/2023]
Abstract
Delivering traditional DNA-damaging anticancer drugs into mitochondria to damage mitochondria is a promising chemotherapy strategy. The impermeability of this mitochondrial inner membrane, however, impedes the delivery of drug molecules that could impact other important biological roles of mitochondria. Herein, the prodrug camptothecin (CPT)-triphenylphosphine (TPP) modified with hyaluronic acid (HA) via electrostatic adsorption (HA/CPT-TPP, HCT) was used to mediate the mitochondrial accumulation of CPT. These nanoparticles (NPs) showed enhanced drug accumulation in cancer cells through tumor targeting. HCT entered acidic lysosomes through endosomal transport, HA was degraded by hyaluronidase (HAase) in acidic lysosomes, and the positively charged CPT-TPP was exposed and accumulated fully in the mitochondria. Subsequently, CPT-TPP significantly disrupted the mitochondrial structure and damaged mitochondrial function, leading to increased reactive oxygen species (ROS) levels and energy depletion. Finally, HCT enhanced lung cancer cell apoptosis via the activation of caspase-3 and caspase-9. Furthermore, greatly increased tumor growth inhibition was observed in nude mice bearing A549 xenograft tumors after the administration of HCT via tail injection. This study demonstrated that the mitochondria-targeted delivery of CPT may be a promising antitumor therapeutic strategy.
Collapse
Affiliation(s)
- Jiacui Xie
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511260, China
| | - He Wang
- Center of Cancer Research, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Qiudi Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiachang Lin
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China; Center of Cancer Research, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Huaying Wen
- The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511260, China
| | - Yingling Miao
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Le Lv
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Dongxue Ruan
- Center of Cancer Research, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiyong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Linghao Qin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Yi Zhou
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
10
|
Zhou Y, Miao Y, Huang Q, Shi W, Xie J, Lin J, Huang P, Yue C, Qin Y, Yu X, Wang H, Qin L, Chen J. A redox-responsive self-assembling COA-4-arm PEG prodrug nanosystem for dual drug delivery suppresses cancer metastasis and drug resistance by downregulating hsp90 expression. Acta Pharm Sin B 2023; 13:3153-3167. [PMID: 37521875 PMCID: PMC10372829 DOI: 10.1016/j.apsb.2022.11.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/09/2022] [Accepted: 11/04/2022] [Indexed: 11/26/2022] Open
Abstract
Metastasis and resistance are main causes to affect the outcome of the current anticancer therapies. Heat shock protein 90 (Hsp90) as an ATP-dependent molecular chaperone takes important role in the tumor metastasis and resistance. Targeting Hsp90 and downregulating its expression show promising in inhibiting tumor metastasis and resistance. In this study, a redox-responsive dual-drug nanocarrier was constructed for the effective delivery of a commonly used chemotherapeutic drug PTX, and a COA-modified 4-arm PEG polymer (4PSC) was synthesized. COA, an active component in oleanolic acid that exerts strong antitumor activity by downregulating Hsp90 expression, was used as a structural and functional element to endow 4PSC with redox responsiveness and Hsp90 inhibitory activity. Our results showed that 4PSC/PTX nanomicelles efficiently delivered PTX and COA to tumor locations without inducing systemic toxicity. By blocking the Hsp90 signaling pathway, 4PSC significantly enhanced the antitumor effect of PTX, inhibiting tumor proliferation and invasiveness as well as chemotherapy-induced resistance in vitro. Remarkable results were further confirmed in vivo with two preclinical tumor models. These findings demonstrate that the COA-modified 4PSC drug delivery nanosystem provides a potential platform for enhancing the efficacy of chemotherapies.
Collapse
Affiliation(s)
- Yi Zhou
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yingling Miao
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Qiudi Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Wenwen Shi
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiacui Xie
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiachang Lin
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Pei Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chengfeng Yue
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
- Center of Cancer Research, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Yuan Qin
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xiyong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology and the State Key Laboratory of Respiratory Disease and the Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - He Wang
- Center of Cancer Research, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Linghao Qin
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jianhai Chen
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
11
|
Gonzalo Ó, Benedi A, Vela L, Anel A, Naval J, Marzo I. Study of the Bcl-2 Interactome by BiFC Reveals Differences in the Activation Mechanism of Bax and Bak. Cells 2023; 12:cells12050800. [PMID: 36899936 PMCID: PMC10000386 DOI: 10.3390/cells12050800] [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: 12/02/2022] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Evasion of apoptosis is one of the hallmarks of cancer cells. Proteins of the Bcl-2 family are key regulators of the intrinsic pathway of apoptosis, and alterations in some of these proteins are frequently found in cancer cells. Permeabilization of the outer mitochondrial membrane, regulated by pro- and antiapoptotic members of the Bcl-2 family of proteins, is essential for the release of apoptogenic factors leading to caspase activation, cell dismantlement, and death. Mitochondrial permeabilization depends on the formation of oligomers of the effector proteins Bax and Bak after an activation event mediated by BH3-only proteins and regulated by antiapoptotic members of the Bcl-2 family. In the present work, we have studied interactions between different members of the Bcl-2 family in living cells via the BiFC technique. Despite the limitations of this technique, present data suggest that native proteins of the Bcl-2 family acting inside living cells establish a complex network of interactions, which would fit nicely into "mixed" models recently proposed by others. Furthermore, our results point to differences in the regulation of Bax and Bak activation by proteins of the antiapoptotic and BH3-only subfamilies. We have also applied the BiFC technique to explore the different molecular models proposed for Bax and Bak oligomerization. Bax and Bak's mutants lacking the BH3 domain were still able to associate and give BiFC signals, suggesting the existence of alternative surfaces of interaction between two Bax or Bak molecules. These results agree with the widely accepted symmetric model for the dimerization of these proteins and also suggest that other regions, different from the α6 helix, could be involved in the oligomerization of BH3-in groove dimers.
Collapse
|
12
|
Richter Syndrome: From Molecular Pathogenesis to Druggable Targets. Cancers (Basel) 2022; 14:cancers14194644. [PMID: 36230566 PMCID: PMC9563287 DOI: 10.3390/cancers14194644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/07/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Richter syndrome (RS) represents the occurrence of an aggressive lymphoma, most commonly diffuse large B-cell lymphoma (DLBCL), in patients with chronic lymphocytic leukemia (CLL). Most cases of RS originate from the direct transformation of CLL, whereas 20% are de novo DLBCL arising as secondary malignancies. Multiple molecular mechanisms contribute to RS pathogenesis. B-cell receptor (BCR) overreactivity to multiple autoantigens is due to frequent stereotyped BCR configuration. Genetic lesions of TP53, CDKN2A, NOTCH1 and c-MYC deregulate DNA damage response, tumor suppression, apoptosis, cell cycle and proliferation. Hyperactivation of Akt and NOTCH1 signaling also plays a role. Altered expression of PD-1/PD-L1 and of other immune checkpoints leads to RS resistance to cytotoxicity exerted by T-cells. The molecular features of RS provide vulnerabilities for therapy. Targeting BCR signaling with noncovalent BTK inhibitors shows encouraging results, as does the combination of BCL2 inhibitors with chemoimmunotherapy. The association of immune checkpoint inhibitors with BCL2 inhibitors and anti-CD20 monoclonal antibodies is explored in early phase clinical trials with promising results. The development of patient-derived xenograft mice models reveals new molecular targets for RS, exemplified by ROR1. Although RS still represents an unmet medical need, understanding its biology is opening new avenues for precision medicine therapy.
Collapse
|
13
|
Manni S, Pesavento M, Spinello Z, Saggin L, Arjomand A, Fregnani A, Quotti Tubi L, Scapinello G, Gurrieri C, Semenzato G, Trentin L, Piazza F. Protein Kinase CK2 represents a new target to boost Ibrutinib and Venetoclax induced cytotoxicity in mantle cell lymphoma. Front Cell Dev Biol 2022; 10:935023. [PMID: 36035991 PMCID: PMC9403710 DOI: 10.3389/fcell.2022.935023] [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: 05/03/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an incurable B cell non-Hodgkin lymphoma, characterized by frequent relapses. In the last decade, the pro-survival pathways related to BCR signaling and Bcl-2 have been considered rational therapeutic targets in B cell derived lymphomas. The BTK inhibitor Ibrutinib and the Bcl-2 inhibitor Venetoclax are emerging as effective drugs for MCL. However, primary and acquired resistance also to these agents may occur. Protein Kinase CK2 is a S/T kinase overexpressed in many solid and blood-derived tumours. CK2 promotes cancer cell growth and clonal expansion, sustaining pivotal survival signaling cascades, such as the ones dependent on AKT, NF-κB, STAT3 and others, counteracting apoptosis through a “non-oncogene” addiction mechanism. We previously showed that CK2 is overexpressed in MCL and regulates the levels of activating phosphorylation on S529 of the NF-κB family member p65/RelA. In the present study, we investigated the effects of CK2 inactivation on MCL cell proliferation, survival and apoptosis and this kinase’s involvement in the BCR and Bcl-2 related signaling. By employing CK2 loss of function MCL cell models, we demonstrated that CK2 sustains BCR signaling (such as BTK, NF-κB and AKT) and the Bcl-2-related Mcl-1 expression. CK2 inactivation enhanced Ibrutinib and Venetoclax-induced cytotoxicity. The demonstration of a CK2-dependent upregulation of pathways that may antagonize the effect of these drugs may offer a novel strategy to overcome primary and secondary resistance.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
- *Correspondence: Sabrina Manni, ; Francesco Piazza,
| | - Maria Pesavento
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Zaira Spinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Lara Saggin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Arash Arjomand
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Anna Fregnani
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Greta Scapinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Carmela Gurrieri
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Livio Trentin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
- *Correspondence: Sabrina Manni, ; Francesco Piazza,
| |
Collapse
|
14
|
Xu W, Zhao T, Chen H, Huang N, Gong H, Zhang J, Yang Y, Li T, Zhang G, Gong C, Yang M, Xiao H. Pan-mTOR inhibitors sensitize the senolytic activity of Navitoclax via mTORC2 inhibition-mediated apoptotic signaling. Biochem Pharmacol 2022; 200:115045. [DOI: 10.1016/j.bcp.2022.115045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 11/30/2022]
|
15
|
Roles and Regulation of BCL-xL in Hematological Malignancies. Int J Mol Sci 2022; 23:ijms23042193. [PMID: 35216310 PMCID: PMC8876520 DOI: 10.3390/ijms23042193] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/20/2022] Open
Abstract
Members of the Bcl-2 family are proteins that play an essential role in the regulation of apoptosis, a crucial process in development and normal physiology in multicellular organisms. The essential mechanism of this family of proteins is given by the role of pro-survival proteins, which inhibit apoptosis by their direct binding with their counterpart, the effector proteins of apoptosis. This family of proteins was named after the typical member Bcl-2, which was named for its discovery and abnormal expression in B-cell lymphomas. Subsequently, the structure of one of its members BCL-xL was described, which allowed one to understand much of the molecular mechanism of this family. Due to its role of BCL-xL in the regulation of cell survival and proliferation, it has been of great interest in its study. Due to this, it is important to research its role regarding the development and progression of human malignancies, especially in hematologic malignancies. Due to its variation in expression in cancer, it has been suggested that BCL-xL can or cannot play a role in cancer depending on the cellular or tissue context. This review discusses recent advances in its transcriptional regulation of BCL-xL, as well as the advances regarding the activities of BCL-xL in hematological malignancies, its possible role as a biomarker, and its possible clinical relevance in these malignancies.
Collapse
|
16
|
Zheng X, Hua S, Zhao H, Gao Z, Cen D. Overexpression of hepatocyte growth factor protects chronic myeloid leukemia cells from apoptosis induced by etoposide. Oncol Lett 2022; 23:122. [PMID: 35261636 DOI: 10.3892/ol.2022.13242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/10/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Xiaojiao Zheng
- Department of Obstetrics and Gynecology, Ningbo First Hospital, Ningbo, Zhejiang 315035, P.R. China
| | - Shixuan Hua
- Department of Laboratory Medicine, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Hang Zhao
- Department of Laboratory Medicine, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Zhou Gao
- Department of Laboratory Medicine, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang 315100, P.R. China
| | - Dong Cen
- Department of Laboratory Medicine, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang 315100, P.R. China
| |
Collapse
|
17
|
Druggable Molecular Pathways in Chronic Lymphocytic Leukemia. Life (Basel) 2022; 12:life12020283. [PMID: 35207569 PMCID: PMC8875960 DOI: 10.3390/life12020283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/08/2022] [Accepted: 02/12/2022] [Indexed: 12/02/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL), the most common type of leukemia in adults, is characterized by a high degree of clinical heterogeneity that is influenced by the disease’s molecular complexity. The genes most frequently affected in CLL cluster into specific biological pathways, including B-cell receptor (BCR) signaling, apoptosis, NF-κB, and NOTCH1 signaling. BCR signaling and the apoptosis pathway have been exploited to design targeted medicines for CLL therapy. Consistently, molecules that selectively inhibit specific BCR components, namely Bruton tyrosine kinase (BTK) and phosphoinositide 3-kinase (PI3K) as well as inhibitors of BCL2, have revolutionized the therapeutic management of CLL patients. Several BTK inhibitors and PI3K inhibitors with different modes of action are currently used or are in development in advanced stage clinical trials. Moreover, the restoration of apoptosis by the BCL2 inhibitor venetoclax offers meaningful clinical activity with a fixed-duration scheme. Inhibitors of the BCR and of BCL2 are able to overcome the chemorefractoriness associated with high-risk genetic features, including TP53 disruption. Other signaling cascades involved in CLL pathogenesis, in particular NOTCH signaling and NF-kB signaling, already provide biomarkers for a precision medicine approach to CLL and may represent potential druggable targets for the future. The aim of the present review is to discuss the druggable pathways of CLL and to provide the biological background of the high efficacy of targeted biological drugs in CLL.
Collapse
|
18
|
Zhang S, Yu H, Li J, Fan J, Chen J. 2-Methoxyestradiol combined with ascorbic acid facilitates the apoptosis of chronic myeloid leukemia cells via the microRNA-223/Fms-like tyrosine kinase 3/phosphatidylinositol-3 kinase/protein kinase B axis. Bioengineered 2022; 13:3470-3485. [PMID: 35068331 PMCID: PMC8973755 DOI: 10.1080/21655979.2021.2024327] [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] [Indexed: 11/02/2022] Open
Abstract
Chronic myeloid leukemia (CML) is a malignant myeloproliferative tumor. 2-Methoxyestradiol (2-ME) is an endogenous estrogen metabolite that shows efficacy in human malignancies. Ascorbic acid (AA) possesses antioxidant activity. This study explored the mechanism of 2-ME combined with AA in the apoptosis of CML cells. Firstly, human CML cell lines were treated with 2-ME and AA. The cell viability, apoptosis, reactive oxygen species (ROS), and mitochondrial membrane potential (MMP) were detected. miR-223 expression in CML cells was detected. In addition, CML cells were transfected with miR-223 inhibitor. The binding relationship between miR-223 and FLT3 was verified. Subsequently, the FLT3 was overexpressed or silenced for the function rescue experiment to confirm the role of FLT3 in CML cell apoptosis. The expression levels of key factors of the PI3K/AKT pathway were detected. Finally, xenograft nude mouse models were established for in vivo verification. 2-ME + AA treatment inhibited CML cell viability and promoted apoptosis, elevated ROS content, and reduced MMP. 2-ME + AA treatment promoted miR-223 expression in CML cells. miR-223 targeted FLT3. Moreover, miR-223 inhibitor or FLT3 overexpression partially annulled the effect of 2-ME + AA on CML cells. 2-ME + AA inhibited the PI3K/AKT pathway via the miR-223/FLT3 axis. Furthermore, 2-ME + AA suppressed CML xenograft growth in mice. Collectively, 2-ME + AA promoted miR-223 expression and suppressed FLT3 and the PI3K/AKT pathway, thereby facilitating the apoptosis of CML cells and inhibiting CML xenograft growth in mice.
Collapse
Affiliation(s)
- Suwei Zhang
- Department of Clinical Laboratory, Shantou Central Hospital, Shantou,Guangdong, China
| | - Hanhui Yu
- Department of Neurosurgery,Shantou Central Hospital, Shantou, Guangdong, China
| | - Jiazhen Li
- Department of Clinical Laboratory, Shantou Central Hospital, Shantou,Guangdong, China
| | - Jingru Fan
- Department of Emergency,Shantou Central Hospital, Shantou, Guangdong, China
| | - Jingchao Chen
- Department of Clinical Laboratory, Shantou Central Hospital, Shantou,Guangdong, China
| |
Collapse
|
19
|
Li G, Li D, Yuan F, Cheng C, Chen L, Wei X. Synergistic effect of chidamide and venetoclax on apoptosis in acute myeloid leukemia cells and its mechanism. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1575. [PMID: 34790781 PMCID: PMC8576699 DOI: 10.21037/atm-21-5066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/16/2021] [Indexed: 01/02/2023]
Abstract
Background Acute myeloid leukemia (AML) is a hematological malignancy with a low remission rate and high recurrence rate. Overexpression of the antiapoptotic protein Bcl-2 is associated with a lower overall survival rate in AML patients. Venetoclax (ABT199) is a selective inhibitor of Bcl-2 that has a significant effect in AML, but single-drug resistance often occurs due to the high expression of Mcl-1 protein. Studies have confirmed that chidamide can downregulate the expression levels of Bcl-2 and Mcl-1 and induce apoptosis. Methods This study aimed to use AML cell lines and primary cells to study the effects of venetoclax and chidamide combination therapy on AML cell apoptosis, the cell cycle, and changes in related signaling pathways in vitro; establish an AML mouse model to observe the efficacy and survival time of combination therapy in vivo; and analyze the drug effects with multi-omics sequencing technology. The changes in gene and protein expression before and after treatment were examined to clarify the molecular mechanism driving the synergistic effect of the two drugs. Results (I) Both venetoclax and chidamide promoted apoptosis in AML cell lines and primary cells in a time- and concentration-dependent manner. The effect was further enhanced when the two drugs were combined, and a synergistic effect was observed (combination index <1). (II) At both the mRNA and protein levels, the expression of Mcl-1 was upregulated by venetoclax and downregulated by chidamide, and the expression of Mcl-1 decreased further after combination treatment. (III) Transcriptome sequencing showed that differentially expressed genes in the combination group compared with the venetoclax monotherapy group were mainly enriched in the PI3K-AKT pathway and JAK2/STAT3 pathway. Moreover, qRT-PCR and Western blot confirmed these results. (IV) The combination therapy group exhibited significantly inhibited disease progression and a prolonged survival time among AML mice. Conclusions Chidamide combined with venetoclax synergistically promoted apoptosis in AML cell lines and primary cells by inhibiting activation of the PI3K/AKT pathway and JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Gangping Li
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongbei Li
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangfang Yuan
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Cheng Cheng
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Chen
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Xudong Wei
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
20
|
Ge L, Lu S, Xu L, Yan H. MYC, BCL2, and BCL6 expression as prognostic indicators in primary central nervous system lymphoma: A systematic review and meta-analysis. Clin Neurol Neurosurg 2021; 208:106838. [PMID: 34339901 DOI: 10.1016/j.clineuro.2021.106838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND We conducted a meta-analysis to comprehensively assess the predictive role of MYC, BCL2, and BCL6 genetic alterations and protein expression in PCNSL for clinical application. METHODS A systematic retrieval was performed on PubMed, Embase, the Cochrane library, Web of Science, Scopus, and 2 Chinese databases. Cohort studies discussing the prognostic impact of MYC, BCl2, or BCL6 genetic alterations or gene expression in PCNSL were selected. The pooled hazard ratio (HR) and median survival ratio (MSR) were calculated. RESULTS 31 studies involving 1739 patients fulfilled our inclusion criteria. MYC expression was significantly associated with short median OS (MSR = 0.62; 95%CI, 0.44-0.88) and PFS (HR = 1.53; 95%CI, 1.06-2.20). No significant association was found between BCL2 expression and OS or PFS (P > 0.05). BCL6 protein positivity was significantly associated with extended median OS (MSR = 1.62; 95%CI, 1.10-2.40). MYC and BCL2 coexpression was significantly associated with short median OS (MSR = 0.61; 95%CI, 0.45-0.84). Subgroup analysis demonstrated that MYC protein positivity remained as a significant indicator for short median OS in studies whose sample size ≥ 45, treatment without WBRT, quality scale score ≥ 7, and positivity threshold set at 40% stratum (MSR < 1 and P < 0.05), but failed to reach a statistically significant difference in the other stratum. CONCLUSIONS MYC expression predicts inferior median OS and PFS in PCNSL. BCL6 protein positivity is associated with a favorable prognosis. The sample size, average age of subjects, WBRT treatment, study quality, and cut-off values for discriminating positive and negative protein expression in IHC may be origins of heterogeneity.
Collapse
Affiliation(s)
- Le Ge
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China.
| | - Shan Lu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China.
| | - Lixia Xu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China.
| | - Hua Yan
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China.
| |
Collapse
|
21
|
Zhong L, Li Y, Xiong L, Wang W, Wu M, Yuan T, Yang W, Tian C, Miao Z, Wang T, Yang S. Small molecules in targeted cancer therapy: advances, challenges, and future perspectives. Signal Transduct Target Ther 2021; 6:201. [PMID: 34054126 PMCID: PMC8165101 DOI: 10.1038/s41392-021-00572-w] [Citation(s) in RCA: 695] [Impact Index Per Article: 173.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/23/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Due to the advantages in efficacy and safety compared with traditional chemotherapy drugs, targeted therapeutic drugs have become mainstream cancer treatments. Since the first tyrosine kinase inhibitor imatinib was approved to enter the market by the US Food and Drug Administration (FDA) in 2001, an increasing number of small-molecule targeted drugs have been developed for the treatment of malignancies. By December 2020, 89 small-molecule targeted antitumor drugs have been approved by the US FDA and the National Medical Products Administration (NMPA) of China. Despite great progress, small-molecule targeted anti-cancer drugs still face many challenges, such as a low response rate and drug resistance. To better promote the development of targeted anti-cancer drugs, we conducted a comprehensive review of small-molecule targeted anti-cancer drugs according to the target classification. We present all the approved drugs as well as important drug candidates in clinical trials for each target, discuss the current challenges, and provide insights and perspectives for the research and development of anti-cancer drugs.
Collapse
Affiliation(s)
- Lei Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, People's Republic of China
| | - Yueshan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Liang Xiong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Wenjing Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ming Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ting Yuan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, People's Republic of China
| | - Wei Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Chenyu Tian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zhuang Miao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Tianqi Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
22
|
Hamilton C, Fox JP, Longley DB, Higgins CA. Therapeutics Targeting the Core Apoptotic Machinery. Cancers (Basel) 2021; 13:cancers13112618. [PMID: 34073507 PMCID: PMC8198123 DOI: 10.3390/cancers13112618] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/09/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Cancer develops when the balance between cell death and cell division in tissues is dysregulated. A key focus of cancer drug discovery is identifying therapeutic agents which will selectively kill and eliminate cancer cells from the body. A number of proteins can prevent the death of cancer cells and developing inhibitors against these proteins to promote cancer cell death is a focus of recent drug discovery efforts. This review aims to summarize the key targets being explored, the drug development approaches being adopted, and the success or limitations of agents currently approved or in clinical development. Abstract Therapeutic targeting of the apoptotic pathways for the treatment of cancer is emerging as a valid and exciting approach in anti-cancer therapeutics. Accumulating evidence demonstrates that cancer cells are typically “addicted” to a small number of anti-apoptotic proteins for their survival, and direct targeting of these proteins could provide valuable approaches for directly killing cancer cells. Several approaches and agents are in clinical development targeting either the intrinsic mitochondrial apoptotic pathway or the extrinsic death receptor mediated pathways. In this review, we discuss the main apoptosis pathways and the key molecular targets which are the subject of several drug development approaches, the clinical development of these agents and the emerging resistance factors and combinatorial treatment approaches for this class of agents with existing and emerging novel targeted anti-cancer therapeutics.
Collapse
|
23
|
Held L, Siu C, Shadman M. Venetoclax as a therapeutic option for the treatment of chronic lymphocytic leukemia: the evidence so far. Expert Opin Pharmacother 2021; 22:655-665. [PMID: 33645381 DOI: 10.1080/14656566.2021.1882421] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Venetoclax, an oral, BCL-2 inhibitor, is approved by the FDA for treatment of CLL in all lines of therapy. Data from landmark studies, including the CLL14 and MURANO trials, demonstrated marked improvement in clinical outcomes compared to chemoimmunotherapy when venetoclax was used in combination with CD20 monoclonal antibodies for fixed treatment duration. AREAS COVERED This article reviews the mechanism of action of venetoclax and discusses how curtailing the BCL signaling pathway undermines CLL pathophysiology. The authors also give their clinical experience with the drug, with emphasis on assessing and managing the risk of venetoclax-associated tumor lysis syndrome (TLS). EXPERT OPINION Venetoclax has positioned itself as one of the primary treatment options for CLL, given the consistent efficacy and deep remissions it has elicited across multiple settings of the disease with a time-limited schedule. Accurate TLS risk evaluation and stringent adherence to the dose-escalation protocols will help optimize patient outcomes. Finally, we expect that current and future studies will (1) ascertain the ideal treatment duration using the minimal residual disease state as a guide and (2) help us understand the optimal role of venetoclax in combination or in sequence with other novel targeted therapies in the treatment of CLL.
Collapse
Affiliation(s)
- Lauren Held
- School of Pharmacy, University of Washington, Seattle, WA, USA.,Hematologic Malignancies, Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Chloe Siu
- School of Pharmacy, University of Washington, Seattle, WA, USA.,Hematologic Malignancies, Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Mazyar Shadman
- School of Pharmacy, University of Washington, Seattle, WA, USA.,Hematologic Malignancies, Seattle Cancer Care Alliance, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
24
|
Wang X, Mak PY, Mu H, Tao W, Rao A, Visweswaran R, Ruvolo V, Pachter JA, Weaver DT, Andreeff M, Xu B, Carter BZ. Combinatorial Inhibition of Focal Adhesion Kinase and BCL-2 Enhances Antileukemia Activity of Venetoclax in Acute Myeloid Leukemia. Mol Cancer Ther 2020; 19:1636-1648. [PMID: 32404407 PMCID: PMC7416436 DOI: 10.1158/1535-7163.mct-19-0841] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/05/2019] [Accepted: 05/07/2020] [Indexed: 01/07/2023]
Abstract
Focal adhesion kinase (FAK) promotes cancer cell growth and metastasis. We previously reported that FAK inhibition by the selective inhibitor VS-4718 exerted antileukemia activities in acute myeloid leukemia (AML). The mechanisms involved, and whether VS-4718 potentiates efficacy of other therapeutic agents, have not been investigated. Resistance to apoptosis inducted by the BCL-2 inhibitor ABT-199 (venetoclax) in AML is mediated by preexisting and ABT-199-induced overexpression of MCL-1 and BCL-XL. We observed that VS-4718 or silencing FAK with siRNA decreased MCL-1 and BCL-XL levels. Importantly, VS-4718 antagonized ABT-199-induced MCL-1 and BCL-XL. VS-4718 markedly synergized with ABT-199 to induce apoptosis in AML cells, including primary AML CD34+ cells and AML cells overexpressing MCL-1 or BCL-XL. In a patient-derived xenograft (PDX) model derived from a patient sample with NPM1/FLT3-ITD/TET2/DNMT3A/WT1 mutations and complex karyotype, VS-4718 statistically significantly reduced leukemia tissue infiltration and extended survival (72 vs. control 36 days, P = 0.0002), and only its combination with ABT-199 effectively decreased systemic leukemia tissue infiltration and circulating blasts, and prolonged survival (65.5 vs. control 36 days, P = 0.0119). Furthermore, the combination decreased NFκB signaling and induced the expression of IFN genes in vivo The combination also markedly extended survival of a second PDX model developed from an aggressive, TP53-mutated complex karyotype AML sample. The data suggest that the combined inhibition of FAK and BCL-2 enhances antileukemia activity in AML at least in part by suppressing MCL-1 and BCL-XL and that this combination may be effective in AML with TP53 and other mutations, and thus benefit patients with high-risk AML.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis
- Biomarkers, Tumor/antagonists & inhibitors
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Proliferation
- Focal Adhesion Kinase 1/antagonists & inhibitors
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Nucleophosmin
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Sulfonamides/pharmacology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Xiangmeng Wang
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Po Yee Mak
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Mu
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wenjing Tao
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Arvind Rao
- The Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ravikumar Visweswaran
- The Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivian Ruvolo
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Bing Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, P.R. China.
- Department of Hematology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, P.R. China
| | - Bing Z Carter
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
25
|
Taylor J, Yeomans AM, Packham G. Targeted inhibition of mRNA translation initiation factors as a novel therapeutic strategy for mature B-cell neoplasms. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:3-25. [PMID: 32924027 PMCID: PMC7116065 DOI: 10.37349/etat.2020.00002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/31/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer development is frequently associated with dysregulation of mRNA translation to enhance both increased global protein synthesis and translation of specific mRNAs encoding oncoproteins. Thus, targeted inhibition of mRNA translation is viewed as a promising new approach for cancer therapy. In this article we review current progress in investigating dysregulation of mRNA translation initiation in mature B-cell neoplasms, focusing on chronic lymphocytic leukemia, follicular lymphoma and diffuse large B-cell lymphoma. We discuss mechanisms and regulation of mRNA translation, potential pathways by which genetic alterations and the tumor microenvironment alters mRNA translation in malignant B cells, preclinical evaluation of drugs targeted against specific eukaryotic initiation factors and current progress towards clinical development. Overall, inhibition of mRNA translation initiation factors is an exciting and promising area for development of novel targeted anti-tumor drugs.
Collapse
Affiliation(s)
- Joe Taylor
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, United Kingdom
| | - Alison M Yeomans
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, United Kingdom
| | - Graham Packham
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, United Kingdom
| |
Collapse
|
26
|
Tarantelli C, Lupia A, Stathis A, Bertoni F. Is There a Role for Dual PI3K/mTOR Inhibitors for Patients Affected with Lymphoma? Int J Mol Sci 2020; 21:E1060. [PMID: 32033478 PMCID: PMC7037719 DOI: 10.3390/ijms21031060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
The activation of the PI3K/AKT/mTOR pathway is a main driver of cell growth, proliferation, survival, and chemoresistance of cancer cells, and, for this reason, represents an attractive target for developing targeted anti-cancer drugs. There are plenty of preclinical data sustaining the anti-tumor activity of dual PI3K/mTOR inhibitors as single agents and in combination in lymphomas. Clinical responses, including complete remissions (especially in follicular lymphoma patients), are also observed in the very few clinical studies performed in patients that are affected by relapsed/refractory lymphomas or chronic lymphocytic leukemia. In this review, we summarize the literature on dual PI3K/mTOR inhibitors focusing on the lymphoma setting, presenting both the three compounds still in clinical development and those with a clinical program stopped or put on hold.
Collapse
Affiliation(s)
- Chiara Tarantelli
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland;
| | - Antonio Lupia
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland;
- Faculty of Biomedical Sciences, USI, 6900 Lugano, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland;
- Oncology Institute of Southern Switzerland, 6500 Bellinzona, Switzerland;
| |
Collapse
|
27
|
Paganelli F, Lonetti A, Anselmi L, Martelli AM, Evangelisti C, Chiarini F. New advances in targeting aberrant signaling pathways in T-cell acute lymphoblastic leukemia. Adv Biol Regul 2019; 74:100649. [PMID: 31523031 DOI: 10.1016/j.jbior.2019.100649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/24/2019] [Accepted: 09/03/2019] [Indexed: 10/26/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disorder characterized by malignant transformation of immature progenitors primed towards T-cell development. Over the past 15 years, advances in the molecular characterization of T-ALL have uncovered oncogenic key drivers and crucial signaling pathways of this disease, opening new chances for the development of novel therapeutic strategies. Currently, T-ALL patients are still treated with aggressive therapies, consisting of high dose multiagent chemotherapy. To minimize and overcome the unfavorable effects of these regimens, it is critical to identify innovative targets and test selective inhibitors of such targets. Major efforts are being made to develop small molecules against deregulated signaling pathways, which sustain T-ALL cell growth, survival, metabolism, and drug-resistance. This review will focus on recent improvements in the understanding of the signaling pathways involved in the pathogenesis of T-ALL and on the challenging opportunities for T-ALL targeted therapies.
Collapse
Affiliation(s)
- Francesca Paganelli
- Institute of Molecular Genetics, Luigi Luca Cavalli-Sforza-CNR National Research Council of Italy, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Annalisa Lonetti
- "Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| | - Laura Anselmi
- Department of Biomedical, Metabolic, and Neural Sciences, Section of Morphology, Signal Transduction Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Camilla Evangelisti
- Institute of Molecular Genetics, Luigi Luca Cavalli-Sforza-CNR National Research Council of Italy, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Chiarini
- Institute of Molecular Genetics, Luigi Luca Cavalli-Sforza-CNR National Research Council of Italy, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| |
Collapse
|