1
|
Norman M, Yamartino K, Gerstein R, Shallis R, Mendez L, Podoltsev N, Stahl M, Eighmy W, Zeidan AM. A review of the isocitrate dehydrogenase inhibitors in management of adult patients with AML and MDS. Expert Rev Hematol 2024:1-13. [PMID: 39474840 DOI: 10.1080/17474086.2024.2422554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/24/2024] [Indexed: 11/09/2024]
Abstract
INTRODUCTION The development of oral therapies impacts the management of acute myeloid leukemia and myelodysplastic syndromes, especially for targetable mutations including IDH1/2. AREAS COVERED We discuss IDH1/2 activity and inhibitor therapy in various settings, including monotherapy, combination therapy with hypomethylating agents, and other approaches. EXPERT OPINION Olutasidenib, enasidenib, and ivosidenib are approved for relapsed AML. Ivosidenib is approved for relapsed MDS and alone or with azacitidine in newly diagnosed AML. However, unanswered questions exist. In newly diagnosed AML, ivosidenib + azacitidine shows a survival benefit compared to azacitidine, but it is unknown whether ivosidenib + azacitidine demonstrates improved survival compared to ivosidenib. Ivosidenib + azacitidine demonstrated a survival benefit not seen with enasidenib + azacitidine. It is unclear whether newly diagnosed AML should be treated with azacitidine + ivosidenib or azacitidine + venetoclax. Azacitidine + venetoclax shows excellent response rates in IDH mutated disease. Retrospective data show low response rates of IDH inhibitor therapy post-venetoclax whereas HMA + venetoclax retains activity post IDH inhibition. The role of IDH inhibition post-transplant is unclear. Single-arm studies show post-transplant maintenance is safe; however, randomized trials are needed. Similarly, IDH inhibitors can be combined with chemotherapy however randomized studies are needed.
Collapse
Affiliation(s)
- Mallory Norman
- Yale New Haven Hospital, Smilow Cancer Center, New Haven, CT, USA
| | | | - Rachel Gerstein
- Yale New Haven Hospital, Smilow Cancer Center, New Haven, CT, USA
| | - Rory Shallis
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Lourdes Mendez
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Nikolai Podoltsev
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Maximilian Stahl
- Department of Medical Oncology, Division of Leukemia, Dana-Farber Cancer Institute, Boston, MA, USA
| | - William Eighmy
- Yale New Haven Hospital, Smilow Cancer Center, New Haven, CT, USA
| | - Amer M Zeidan
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
2
|
Dinghuan W, Yi K, Jianzhi T, Wenfei W, Chunlin W, Anling H, Zhixu H, Ben-David Y, Sheng L, Xiaoyan Y, Xiao X. A novel iheyamine A derivative L42 suppresses acute myeloid leukemia via dual regulation of the PI3K/AKT/FOXO3a axis and TNF signaling pathway. Biomed Pharmacother 2024; 177:117071. [PMID: 38981243 DOI: 10.1016/j.biopha.2024.117071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/22/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024] Open
Abstract
Acute myeloid leukemia (AML) is one of the most common hematopoietic malignancies and the development of new drugs is crucial for the treatment of this lethal disease. Iheyamine A is a nonmonoterpenoid azepinoindole alkaloid from the ascidian Polycitorella sp., and its anticancer mechanism has not been investigated in leukemias. Herein, we showed the significant antileukemic activity of L42 in AML cell lines HEL, HL-60 and THP-1. The IC50 values were 0.466±0.099 µM, 0.356±0.023 µM, 0.475±0.084 µM in the HEL, HL-60 and THP-1 cell lines, respectively, which were lower than the IC50 (2.594±0.271 µM) in the normal liver cell line HL-7702. Furthermore, L42 significantly inhibited the growth of peripheral blood mononuclear cells (PBMCs) from an AML patient. In vivo, L42 effectively suppressed leukemia progression in a mouse model induced by Friend murine leukemia virus (F-MuLV). Mechanistically, we showed that L42 induced cell cycle arrest and apoptosis in leukemia cell lines. RNA sequencing analysis of L42-treated THP-1 cells revealed that the differentially expressed genes (DEGs) were enriched in the cell cycle and apoptosis and predominantly enriched in the PI3K/AKT pathway. Accordingly, L42 decreased the expression of the phospho-PI3K (p85), phospho-AKT and phospho-FOXO3a. Docking and CETSA analysis indicated that L42 bound to the PI3K isoform p110α (PIK3CA), which was implicated in the suppression of the PI3K/AKT pathway. L42 was also shown to initiate the TNF signaling-mediated apoptosis. Moreover, L42 exhibited stronger anti-leukemia activity and sensitivity in IDH2-mutant HEL cells than in IDH2-wild-type control. In conclusion, L42 effectively suppresses cell proliferation and triggers apoptosis in AML cell lines in part through inhibition of the PI3K/AKT signaling pathway to restore FOXO3a expression and activation of the TNF signaling pathway. Thus, the iheyamine A derivative L42 represents a novel candidate for AML therapy.
Collapse
Affiliation(s)
- Wang Dinghuan
- Department of Pediatrics, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, PR China; State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou 550014, PR China
| | - Kuang Yi
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550014, PR China
| | - Tian Jianzhi
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou 550014, PR China
| | - Wei Wenfei
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou 550014, PR China
| | - Wang Chunlin
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou 550014, PR China
| | - Hu Anling
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou 550014, PR China
| | - He Zhixu
- Department of Pediatrics, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, PR China
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou 550014, PR China.
| | - Liu Sheng
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou 550014, PR China.
| | - Yang Xiaoyan
- Department of Pediatrics, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, PR China.
| | - Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang, Guizhou 550014, PR China.
| |
Collapse
|
3
|
Ali A, Phan A, Vaikari V, Park M, Pospiech M, Chu R, Meng Y, MacKay JA, Alachkar H. FLT3/CD99 Bispecific Antibody-Based Nanoparticles for Acute Myeloid Leukemia. CANCER RESEARCH COMMUNICATIONS 2024; 4:1946-1962. [PMID: 39007347 PMCID: PMC11305399 DOI: 10.1158/2767-9764.crc-24-0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/22/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Cluster of differentiation 99 (CD99) is a receptor that is significantly upregulated in acute myeloid leukemia (AML). FMS-like tyrosine kinase 3 internal tandem duplication mutation in AML (FLT3-ITD AML) exhibits even higher levels of CD99 expression. Our group previously employed a novel peptide platform technology called elastin-like polypeptides and fused it with single-chain antibodies capable of binding to FLT3 (FLT3-A192) or CD99 (CD99-A192). Targeting either FLT3 or CD99 using FLT3-A192 or CD99-A192 led to AML cell death and reduced leukemia burden in AML mouse models. Here, we report on the development of a novel Co-Assembled construct that is capable of binding to both CD99 and FLT3 and the antileukemia activity of the bispecific construct in FLT3-ITD AML preclinical models. This dual-targeting Co-Assembled formulation exhibits cytotoxic effects on AML cells (AML cell lines and primary blasts) and reduced leukemia burden and prolonged survival in FLT3-ITD AML mouse models. Altogether, this study demonstrates the potential of an innovative therapeutic strategy that targets both FLT3 and CD99 in FLT3-ITD AML. SIGNIFICANCE This study investigates a dual-targeting strategy in acute myeloid leukemia (AML), focusing on FLT3 and CD99. The approach demonstrates enhanced therapeutic potential, presenting a novel option for AML treatment.
Collapse
Affiliation(s)
- Atham Ali
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Alvin Phan
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Vijaya Vaikari
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Mincheol Park
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Mateusz Pospiech
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Ryan Chu
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - Yiting Meng
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
| | - J. Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, University of Southern California, Los Angeles, California.
- Department of Ophthalmology, USC Roski Eye Institute, USC Keck School of Medicine, University of Southern California, Los Angeles, California.
- Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California.
| | - Houda Alachkar
- Department of Clinical Pharmacy, USC School of Pharmacy, University of Southern California, Los Angeles, California.
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California.
| |
Collapse
|
4
|
Kowalczyk A, Zarychta J, Lejman M, Latoch E, Zawitkowska J. Clinical Implications of Isocitrate Dehydrogenase Mutations and Targeted Treatment of Acute Myeloid Leukemia with Mutant Isocitrate Dehydrogenase Inhibitors-Recent Advances, Challenges and Future Prospects. Int J Mol Sci 2024; 25:7916. [PMID: 39063158 PMCID: PMC11276768 DOI: 10.3390/ijms25147916] [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: 06/21/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Despite the better understanding of the molecular mechanisms contributing to the pathogenesis of acute myeloid leukemia (AML) and improved patient survival in recent years, AML therapy still remains a clinical challenge. For this reason, it is important to search for new therapies that will enable the achievement of remission. Recently, the Food and Drug Administration approved three mutant IDH (mIDH) inhibitors for the treatment of AML. However, the use of mIDH inhibitors in monotherapy usually leads to the development of resistance and the subsequent recurrence of the cancer, despite the initial effectiveness of the therapy. A complete understanding of the mechanisms by which IDH mutations influence the development of leukemia, as well as the processes that enable resistance to mIDH inhibitors, may significantly improve the efficacy of this therapy through the use of an appropriate synergistic approach. The aim of this literature review is to present the role of IDH1/IDH2 mutations in the pathogenesis of AML and the results of clinical trials using mIDH1/IDH2 inhibitors in AML and to discuss the challenges related to the use of mIDH1/IDH2 inhibitors in practice and future prospects related to the potential methods of overcoming resistance to these agents.
Collapse
Affiliation(s)
- Adrian Kowalczyk
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (J.Z.)
| | - Julia Zarychta
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (J.Z.)
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Eryk Latoch
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, 15-274 Bialystok, Poland;
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
5
|
Carullo G, Rossi S, Giudice V, Pezzotta A, Chianese U, Scala P, Carbone S, Fontana A, Panzeca G, Pasquini S, Contri C, Gemma S, Ramunno A, Saponara S, Galvani F, Lodola A, Mor M, Benedetti R, Selleri C, Varani K, Butini S, Altucci L, Vincenzi F, Pistocchi A, Campiani G. Development of Epigenetic Modifiers with Therapeutic Potential in FMS-Related Tyrosine Kinase 3/Internal Tandem Duplication (FLT3/ITD) Acute Myeloid Leukemia and Other Blood Malignancies. ACS Pharmacol Transl Sci 2024; 7:2125-2142. [PMID: 39022363 PMCID: PMC11249625 DOI: 10.1021/acsptsci.4c00208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/04/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024]
Abstract
Blood cancers encompass a group of diseases affecting the blood, bone marrow, or lymphatic system, representing the fourth most commonly diagnosed cancer worldwide. Leukemias are characterized by the dysregulated proliferation of myeloid and lymphoid cells with different rates of progression (acute or chronic). Among the chronic forms, hairy cell leukemia (HCL) is a rare disease, and no drugs have been approved to date. However, acute myeloid leukemia (AML) is one of the most aggressive malignancies, with a low survival rate, especially in cases with FLT3-ITD mutations. Epigenetic modifications have emerged as promising strategies for the treatment of blood cancers. Epigenetic modulators, such as histone deacetylase (HDAC) inhibitors, are increasingly used for targeted cancer therapy. New hydroxamic acid derivatives, preferentially inhibiting HDAC6 (5a-q), were developed and their efficacy was investigated in different blood cancers, including multiple myeloma (MM), HCL, and AML, pointing out their pro-apoptotic effect as the mechanism of cell death. Among the inhibitors described, 5c, 5g, and 5h were able to rescue the hematopoietic phenotype in vivo using the FLT3-ITD zebrafish model of AML. 5c (leuxinostat) proved its efficacy in cells from FLT3-ITD AML patients, promoting marked acetylation of α-tubulin compared to histone H3, thereby confirming HDAC6 as a preferential target for this new class of hydroxamic acid derivatives at the tested doses.
Collapse
Affiliation(s)
- Gabriele Carullo
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Sara Rossi
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Valentina Giudice
- Department
of Medicine, Surgery, Dentistry “Scuola Medica Salernitana”, University of Salerno, Via S. Allende, Baronissi, SA 84081, Italy
| | - Alex Pezzotta
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, LITA, Fratelli Cervi 93, Segrate, MI 20054, Italy
| | - Ugo Chianese
- Department
of Precision Medicine, University of Campania
Luigi Vanvitelli, Via de Crecchio 7, Naples 80138, Italy
| | - Pasqualina Scala
- Department
of Medicine, Surgery, Dentistry “Scuola Medica Salernitana”, University of Salerno, Via S. Allende, Baronissi, SA 84081, Italy
| | - Sabrina Carbone
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, LITA, Fratelli Cervi 93, Segrate, MI 20054, Italy
| | - Anna Fontana
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Giovanna Panzeca
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Silvia Pasquini
- Department
of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Borsari 46, Ferrara 44121, Italy
| | - Chiara Contri
- Department
of Translational Medicine, University of
Ferrara, Via Borsari 46, Ferrara 44121, Italy
| | - Sandra Gemma
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Anna Ramunno
- Department
of Pharmacy, University of Salerno, Giovanni Paolo II, 132, Fisciano, SA 84084, Italy
| | - Simona Saponara
- Department
of Life Sciences, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Francesca Galvani
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Alessio Lodola
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Marco Mor
- Department
of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, Parma 43124, Italy
| | - Rosaria Benedetti
- Department
of Precision Medicine, University of Campania
Luigi Vanvitelli, Via de Crecchio 7, Naples 80138, Italy
- Program
of Medical Epigenetics, Vanvitelli Hospital, Naples 80138, Italy
| | - Carmine Selleri
- Department
of Medicine, Surgery, Dentistry “Scuola Medica Salernitana”, University of Salerno, Via S. Allende, Baronissi, SA 84081, Italy
| | - Katia Varani
- Department
of Translational Medicine, University of
Ferrara, Via Borsari 46, Ferrara 44121, Italy
| | - Stefania Butini
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Lucia Altucci
- Department
of Precision Medicine, University of Campania
Luigi Vanvitelli, Via de Crecchio 7, Naples 80138, Italy
- Program
of Medical Epigenetics, Vanvitelli Hospital, Naples 80138, Italy
- Biogem
Institute of Molecular and Genetic Biology, Ariano Irpino 83031, Italy
| | - Fabrizio Vincenzi
- Department
of Translational Medicine, University of
Ferrara, Via Borsari 46, Ferrara 44121, Italy
| | - Anna Pistocchi
- Department
of Medical Biotechnology and Translational Medicine, University of Milan, LITA, Fratelli Cervi 93, Segrate, MI 20054, Italy
| | - Giuseppe Campiani
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena 53100, Italy
- Bioinformatics
Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-7346, Iran
| |
Collapse
|
6
|
Tecik M, Adan A. Emerging DNA Methylome Targets in FLT3-ITD-Positive Acute Myeloid Leukemia: Combination Therapy with Clinically Approved FLT3 Inhibitors. Curr Treat Options Oncol 2024; 25:719-751. [PMID: 38696033 PMCID: PMC11222205 DOI: 10.1007/s11864-024-01202-7] [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] [Accepted: 04/01/2024] [Indexed: 07/04/2024]
Abstract
OPINION STATEMENT The internal tandem duplication (ITD) mutation of the FMS-like receptor tyrosine kinase 3 (FLT3-ITD) is the most common mutation observed in approximately 30% of acute myeloid leukemia (AML) patients. It represents poor prognosis due to continuous activation of downstream growth-promoting signaling pathways such as STAT5 and PI3K/AKT. Hence, FLT3 is considered an attractive druggable target; selective small FLT3 inhibitors (FLT3Is), such as midostaurin and quizartinib, have been clinically approved. However, patients possess generally poor remission rates and acquired resistance when FLT3I used alone. Various factors in patients could cause these adverse effects including altered epigenetic regulation, causing mainly abnormal gene expression patterns. Epigenetic modifications are required for hematopoietic stem cell (HSC) self-renewal and differentiation; however, critical driver mutations have been identified in genes controlling DNA methylation (such as DNMT3A, TET2, IDH1/2). These regulators cause leukemia pathogenesis and affect disease diagnosis and prognosis when they co-occur with FLT3-ITD mutation. Therefore, understanding the role of different epigenetic alterations in FLT3-ITD AML pathogenesis and how they modulate FLT3I's activity is important to rationalize combinational treatment approaches including FLT3Is and modulators of methylation regulators or pathways. Data from ongoing pre-clinical and clinical studies will further precisely define the potential use of epigenetic therapy together with FLT3Is especially after characterized patients' mutational status in terms of FLT3 and DNA methlome regulators.
Collapse
Affiliation(s)
- Melisa Tecik
- Bioengineering Program, Graduate School of Engineering and Science, Abdullah Gul University, Kayseri, Turkey
| | - Aysun Adan
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, Turkey.
| |
Collapse
|
7
|
Yang J, Friedman R. Synergy and antagonism between azacitidine and FLT3 inhibitors. Comput Biol Med 2024; 169:107889. [PMID: 38199214 DOI: 10.1016/j.compbiomed.2023.107889] [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: 09/27/2023] [Revised: 12/05/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024]
Abstract
Synergetic interactions between drugs can make a drug combination more effective. Alternatively, they may allow to use lower concentrations and thus avoid toxicities or side effects that not only cause discomfort but might also reduce the overall survival. Here, we studied whether synergy exists between agents that are used for treatment of acute myeloid leukaemia (AML). Azacitidine is a demethylation agent that is used in the treatment of AML patients that are unfit for aggressive chemotherapy. An activating mutation in the FLT3 gene is common in AML patients and in the absence of specific treatment makes prognosis worse. FLT3 inhibitors may be used in such cases. We sought to determine whether combination of azacitidine with a FLT3 inhibitor (gilteritinib, quizartinib, LT-850-166, FN-1501 or FF-10101) displayed synergy or antagonism. To this end, we calculated dose-response matrices of these drug combinations from experiments in human AML cells and subsequently analysed the data using a novel consensus scoring algorithm. The results show that combinations that involved non-covalent FLT3 inhibitors, including the two clinically approved drugs gilteritinib and quizartinib were antagonistic. On the other hand combinations with the covalent inhibitor FF-10101 had some range of concentrations where synergy was observed.
Collapse
Affiliation(s)
- Jingmei Yang
- Department of Chemistry and Biomedical Science, Linnaeus University, Kalmar, SE-39231, Sweden
| | - Ran Friedman
- Department of Chemistry and Biomedical Science, Linnaeus University, Kalmar, SE-39231, Sweden.
| |
Collapse
|
8
|
Abdel-Aziz AK, Dokla EME, Saadeldin MK. FLT3 inhibitors and novel therapeutic strategies to reverse AML resistance: An updated comprehensive review. Crit Rev Oncol Hematol 2023; 191:104139. [PMID: 37717880 DOI: 10.1016/j.critrevonc.2023.104139] [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: 04/01/2023] [Revised: 08/20/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) mutations occur in almost 30% of acute myeloid leukemia (AML) patients. Despite the initial clinical efficacy of FLT3 inhibitors, many treated AML patients with mutated FLT3 eventually relapse. This review critically discusses the opportunities and challenges of FLT3-targeted therapies and sheds light on their drug interactions as well as potential biomarkers. Furthermore, we focus on the molecular mechanisms underlying the resistance of FLT3 internal tandem duplication (FLT3-ITD) AMLs to FLT3 inhibitors alongside novel therapeutic strategies to reverse resistance. Notably, dynamic heterogeneous patterns of clonal selection and evolution contribute to the resistance of FLT3-ITD AMLs to FLT3 inhibitors. Ongoing preclinical research and clinical trials are actively directed towards devising rational "personalized" or "patient-tailored" combinatorial therapeutic regimens to effectively treat patients with FLT3 mutated AML.
Collapse
Affiliation(s)
- Amal Kamal Abdel-Aziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt; Smart Health Initiative, Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia.
| | - Eman M E Dokla
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo 11566, Egypt
| | - Mona Kamal Saadeldin
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Leahy Drive, Notre Dame, IN 46556, USA
| |
Collapse
|
9
|
Negotei C, Colita A, Mitu I, Lupu AR, Lapadat ME, Popovici CE, Crainicu M, Stanca O, Berbec NM. A Review of FLT3 Kinase Inhibitors in AML. J Clin Med 2023; 12:6429. [PMID: 37892567 PMCID: PMC10607239 DOI: 10.3390/jcm12206429] [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: 08/29/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive illness distinguished by the accumulation of abnormal hematopoietic precursors in both the bone marrow and peripheral blood. The prevalence of FLT3 gene mutations is high and escalates the probability of relapse and mortality. The survival rates for AML patients, particularly those over 65, are low. FLT3 mutation screening at diagnosis is mandatory, and FLT3 inhibitors are crucial in treating AML patients with mutations. There are two categories of FLT3 mutations: FLT3-ITD located in the juxtamembrane domain and FLT3-TKD in the tyrosine kinase domain. FLT3-ITD is the most common type, affecting nearly a quarter of patients, whereas FLT3-TKD only affects 6-8% of patients. FLT3 inhibitors are now crucial in treating AML patients with FLT3 mutations. When dealing with FLT3-mutated AML, the recommended course of treatment typically involves chemotherapy and midostaurin, followed by allogeneic hematopoietic cell transplantation (HCT) to maximize the likelihood of success. Maintenance therapy can lower the risk of relapse, and gilteritinib is a better option than salvage chemotherapy for relapsed or refractory cases. Clinical trials for new or combined therapies are the most effective approach. This review discusses treatment options for patients with FLT3-mutated AML, including induction chemotherapy and options for relapsed or refractory disease. Additional treatment options may become available as more studies are conducted based on the patient's condition and susceptibility.
Collapse
Affiliation(s)
- Cristina Negotei
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Clinic of Hematology, Coltea Clinical Hospital, 030171 Bucharest, Romania
| | - Andrei Colita
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Clinic of Hematology, Coltea Clinical Hospital, 030171 Bucharest, Romania
| | - Iuliana Mitu
- Clinic of Hematology, Coltea Clinical Hospital, 030171 Bucharest, Romania
| | - Anca Roxana Lupu
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mihai-Emilian Lapadat
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Clinic of Hematology, Coltea Clinical Hospital, 030171 Bucharest, Romania
| | - Constanta Elena Popovici
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Clinic of Hematology, Coltea Clinical Hospital, 030171 Bucharest, Romania
| | - Madalina Crainicu
- Clinic of Hematology, Coltea Clinical Hospital, 030171 Bucharest, Romania
| | - Oana Stanca
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Clinic of Hematology, Coltea Clinical Hospital, 030171 Bucharest, Romania
| | - Nicoleta Mariana Berbec
- Department of Hematology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Clinic of Hematology, Coltea Clinical Hospital, 030171 Bucharest, Romania
| |
Collapse
|
10
|
Wu RH, Zhu CY, Yu PH, Ma Y, Hussain L, Naranmandura H, Wang QQ. The landscape of novel strategies for acute myeloid leukemia treatment: Therapeutic trends, challenges, and future directions. Toxicol Appl Pharmacol 2023; 473:116585. [PMID: 37302559 DOI: 10.1016/j.taap.2023.116585] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023]
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous subtype of hematological malignancies with a wide spectrum of cytogenetic and molecular abnormalities, which makes it difficult to manage and cure. Along with the deeper understanding of the molecular mechanisms underlying AML pathogenesis, a large cohort of novel targeted therapeutic approaches has emerged, which considerably expands the medical options and changes the therapeutic landscape of AML. Despite that, resistant and refractory cases caused by genomic mutations or bypass signalling activation remain a great challenge. Therefore, discovery of novel treatment targets, optimization of combination strategies, and development of efficient therapeutics are urgently required. This review provides a detailed and comprehensive discussion on the advantages and limitations of targeted therapies as a single agent or in combination with others. Furthermore, the innovative therapeutic approaches including hyperthermia, monoclonal antibody-based therapy, and CAR-T cell therapy are also introduced, which may provide safe and viable options for the treatment of patients with AML.
Collapse
Affiliation(s)
- Ri Han Wu
- College of Life Sciences, Changchun Normal University, Changchun 130032, China
| | - Chen Ying Zhu
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Pei Han Yu
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yafang Ma
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liaqat Hussain
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Hua Naranmandura
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Qian Qian Wang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
11
|
Nguyen A, Nguyen A, Dada OT, Desai PD, Ricci JC, Godbole NB, Pierre K, Lucke-Wold B. Leptomeningeal Metastasis: A Review of the Pathophysiology, Diagnostic Methodology, and Therapeutic Landscape. Curr Oncol 2023; 30:5906-5931. [PMID: 37366925 DOI: 10.3390/curroncol30060442] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
The present review aimed to establish an understanding of the pathophysiology of leptomeningeal disease as it relates to late-stage development among different cancer types. For our purposes, the focused metastatic malignancies include breast cancer, lung cancer, melanoma, primary central nervous system tumors, and hematologic cancers (lymphoma, leukemia, and multiple myeloma). Of note, our discussion was limited to cancer-specific leptomeningeal metastases secondary to the aforementioned primary cancers. LMD mechanisms secondary to non-cancerous pathologies, such as infection or inflammation of the leptomeningeal layer, were excluded from our scope of review. Furthermore, we intended to characterize general leptomeningeal disease, including the specific anatomical infiltration process/area, CSF dissemination, manifesting clinical symptoms in patients afflicted with the disease, detection mechanisms, imaging modalities, and treatment therapies (both preclinical and clinical). Of these parameters, leptomeningeal disease across different primary cancers shares several features. Pathophysiology regarding the development of CNS involvement within the mentioned cancer subtypes is similar in nature and progression of disease. Consequently, detection of leptomeningeal disease, regardless of cancer type, employs several of the same techniques. Cerebrospinal fluid analysis in combination with varied imaging (CT, MRI, and PET-CT) has been noted in the current literature as the gold standard in the diagnosis of leptomeningeal metastasis. Treatment options for the disease are both varied and currently in development, given the rarity of these cases. Our review details the differences in leptomeningeal disease as they pertain through the lens of several different cancer subtypes in an effort to highlight the current state of targeted therapy, the potential shortcomings in treatment, and the direction of preclinical and clinical treatments in the future. As there is a lack of comprehensive reviews that seek to characterize leptomeningeal metastasis from various solid and hematologic cancers altogether, the authors intended to highlight not only the overlapping mechanisms but also the distinct patterning of disease detection and progression as a means to uniquely treat each metastasis type. The scarcity of LMD cases poses a barrier to more robust evaluations of this pathology. However, as treatments for primary cancers have improved over time, so has the incidence of LMD. The increase in diagnosed cases only represents a small fraction of LMD-afflicted patients. More often than not, LMD is determined upon autopsy. The motivation behind this review stems from the increased capacity to study LMD in spite of scarcity or poor patient prognosis. In vitro analysis of leptomeningeal cancer cells has allowed researchers to approach this disease at the level of cancer subtypes and markers. We ultimately hope to facilitate the clinical translation of LMD research through our discourse.
Collapse
Affiliation(s)
- Andrew Nguyen
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Alexander Nguyen
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | - Persis D Desai
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jacob C Ricci
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Nikhil B Godbole
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Kevin Pierre
- Department of Radiology, University of Florida, Gainesville, FL 32610, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
12
|
Perrone S, Ottone T, Zhdanovskaya N, Molica M. How acute myeloid leukemia (AML) escapes from FMS-related tyrosine kinase 3 (FLT3) inhibitors? Still an overrated complication? CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:223-238. [PMID: 37457126 PMCID: PMC10344728 DOI: 10.20517/cdr.2022.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/21/2023] [Accepted: 04/11/2023] [Indexed: 07/18/2023]
Abstract
FMS-related tyrosine kinase 3 (FLT3) mutations, present in about 25%-30% of acute myeloid leukemia (AML) patients, constitute one of the most frequently detected mutations in these patients. The binding of FLT3L to FLT3 activates the phosphatidylinositol 3-kinase (PI3K) and RAS pathways, producing increased cell proliferation and the inhibition of apoptosis. Two types of FLT3 mutations exist: FLT3-ITD and FLT3-TKD (point mutations in D835 and I836 or deletion of codon I836). A class of drugs, tyrosine-kinase inhibitors (TKI), targeting mutated FLT3, is already available with 1st and 2nd generation molecules, but only midostaurin and gilteritinib are currently approved. However, the emergence of resistance or the selection of clones not responding to FLT3 inhibitors has become an important clinical dilemma, as the duration of clinical responses is generally limited to a few months. This review analyzes the insights into mechanisms of resistance to TKI and poses a particular view on the clinical relevance of this phenomenon. Has resistance been overlooked? Indeed, FLT3 inhibitors have significantly contributed to reducing the negative impact of FLT3 mutations on the prognosis of AML patients who are no longer considered at high risk by the European LeukemiaNet (ELN) 2022. Finally, several ongoing efforts to overcome resistance to FLT3-inhibitors will be presented: new generation FLT3 inhibitors in monotherapy or combined with standard chemotherapy, hypomethylating drugs, or IDH1/2 inhibitors, Bcl2 inhibitors; novel anti-human FLT3 monoclonal antibodies (e.g., FLT3/CD3 bispecific antibodies); FLT3-CAR T-cells; CDK4/6 kinase inhibitor (e.g., palbociclib).
Collapse
Affiliation(s)
- Salvatore Perrone
- Hematology, Polo Universitario Pontino, S.M. Goretti Hospital, Latina 04100, Italy
| | - Tiziana Ottone
- Department of Biomedicine and Prevention, the University of Rome “Tor Vergata”, Rome 00100 Italy
- Neuro-Oncohematology, Santa Lucia Foundation, I.R.C.C.S., Rome 00100, Italy
| | - Nadezda Zhdanovskaya
- Hematology, Department of Translational and Precision Medicine, Sapienza University, Rome 00161, Italy
| | - Matteo Molica
- Hematology Unit, S. Eugenio Hospital, ASL Roma 2, Rome 00144, Italy
| |
Collapse
|
13
|
Venugopal S, Xie Z, Zeidan AM. An overview of novel therapies in advanced clinical testing for acute myeloid leukemia. Expert Rev Hematol 2023; 16:109-119. [PMID: 36718500 DOI: 10.1080/17474086.2023.2174521] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION The past decade has seen a sea change in the AML landscape with vastly improved cognizance of molecular pathogenesis, clonal evolution, and importance of measurable residual disease. Since 2017, the therapeutic armamentarium of AML has considerably expanded with the approval of midostaurin, enasidenib, ivosidenib, gilteritinib, and venetoclax in combination with hypomethylating agents and others. Nevertheless, relapse and treatment refractoriness remain the insurmountable challenges in AML therapy. This has galvanized the leukemic research community leading to the discovery and development of agents that specifically target gene mutations, molecularly agnostic therapies that exploit immune environment, apoptotic pathways, leukemic cell surface antigens and so forth. AREAS COVERED This article provides an overview of the pathophysiology of AML in the context of non-cellular immune and molecularly targeted and agnostic therapies that are in clinical trial development in AML. EXPERT OPINION Ever growing understanding of the molecular pathogenesis and metabolomics in AML has allowed the researchers to identify targets directed at specific genes and metabolic pathways. As a result, AML therapy is constantly evolving and so are the escape mechanisms leading to disease relapse. Therefore, it is of paramount importance to sequentially evaluate the patient during AML treatment and intervene at the right time.
Collapse
Affiliation(s)
- Sangeetha Venugopal
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Zhuoer Xie
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, FL, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
14
|
Garciaz S, Hospital MA. FMS-Like Tyrosine Kinase 3 Inhibitors in the Treatment of Acute Myeloid Leukemia: An Update on the Emerging Evidence and Safety Profile. Onco Targets Ther 2023; 16:31-45. [PMID: 36698434 PMCID: PMC9869913 DOI: 10.2147/ott.s236740] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) is one of the most frequently mutated genes in acute myeloid leukemia (AML). Approximately 30% of the adult cases harbor an internal tandem duplication (FLT3-ITD) and 5-10% a tyrosine kinase domain (TKD) amino acid substitution (FLT3-TKD). The treatment paradigm of AML patients harboring FLT3 mutations (30%) has been modified by the discovery of tyrosine kinase inhibitors. First- and second-generation inhibitors classify FLT3 inhibitors according to FLT3 specificity: first-generation FLT3 inhibitors include sorafenib and midostaurin and second-generation inhibitors are represented by quizartinib, gilteritinib and crenolanib, among others. Activity of these inhibitors depends on their mechanism of receptor binding (active vs inactive conformation) and efficacy against the FLT3-ITD and -TKD mutations (type 1 inhibitors are active both on FLT3-ITD and TKD, whereas type 2 inhibitors are active only on FLT3-ITD). The FLT3 inhibitors sorafenib, midostaurin, quizartinib and gilteritinib have been tested in monotherapy in several settings including refractory or relapsed AML (R/R AML), post-transplant maintenance as well as in combination with intensive chemotherapy (ICT) or non-intensity regimens. The results of published randomized studies support the use of sorafenib in a post-transplant setting (SORMAIN trial), midostaurin in combination with ICT based (RATIFY trial) and gilteritinib for R/R AML (ADMIRAL trial). Gilteritinib in combination with hypomethylating agent as well as quizartinib are not supported by solid randomized trial results for their use in FLT3-mutated AML patients.
Collapse
Affiliation(s)
- Sylvain Garciaz
- Department of Hematology, Institut Paoli-Calmettes, Aix-Marseille Université, Centre National de la Recherche Scientifique, UMR7258, Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Marseille, France,Correspondence: Sylvain Garciaz, Institut Paoli-Calmettes, Hematology Department, 232 Bd Sainte-Marguerite, Marseille, 13009, France, Tel + 33 4 91 22 37 54, Fax + 33 4 91 22 30 63, Email
| | - Marie-Anne Hospital
- Department of Hematology, Institut Paoli-Calmettes, Aix-Marseille Université, Centre National de la Recherche Scientifique, UMR7258, Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Marseille, France
| |
Collapse
|
15
|
Min HY, Lee HY. Molecular targeted therapy for anticancer treatment. Exp Mol Med 2022; 54:1670-1694. [PMID: 36224343 PMCID: PMC9636149 DOI: 10.1038/s12276-022-00864-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 02/07/2023] Open
Abstract
Since the initial clinical approval in the late 1990s and remarkable anticancer effects for certain types of cancer, molecular targeted therapy utilizing small molecule agents or therapeutic monoclonal antibodies acting as signal transduction inhibitors has served as a fundamental backbone in precision medicine for cancer treatment. These approaches are now used clinically as first-line therapy for various types of human cancers. Compared to conventional chemotherapy, targeted therapeutic agents have efficient anticancer effects with fewer side effects. However, the emergence of drug resistance is a major drawback of molecular targeted therapy, and several strategies have been attempted to improve therapeutic efficacy by overcoming such resistance. Herein, we summarize current knowledge regarding several targeted therapeutic agents, including classification, a brief biology of target kinases, mechanisms of action, examples of clinically used targeted therapy, and perspectives for future development.
Collapse
Affiliation(s)
- Hye-Young Min
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ho-Young Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Lama R, Xu C, Galster SL, Querol-García J, Portwood S, Mavis CK, Ruiz FM, Martin D, Wu J, Giorgi MC, Bargonetti J, Wang ES, Hernandez-Ilizaliturri FJ, Koudelka GB, Chemler SR, Muñoz IG, Wang X. Small molecule MMRi62 targets MDM4 for degradation and induces leukemic cell apoptosis regardless of p53 status. Front Oncol 2022; 12:933446. [PMID: 35992795 PMCID: PMC9389462 DOI: 10.3389/fonc.2022.933446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/11/2022] [Indexed: 12/15/2022] Open
Abstract
MDM2 and MDM4 proteins are key negative regulators of tumor suppressor p53. MDM2 and MDM4 interact via their RING domains and form a heterodimer polyubiquitin E3 ligase essential for p53 degradation. MDM4 also forms heterodimer E3 ligases with MDM2 isoforms that lack p53-binding domains, which regulate p53 and MDM4 stability. We are working to identify small-molecule inhibitors targeting the RING domain of MDM2-MDM4 (MMRi) that can inactivate the total oncogenic activity of MDM2-MDM4 heterodimers. Here, we describe the identification and characterization of MMRi62 as an MDM4-degrader and apoptosis inducer in leukemia cells. Biochemically, in our experiments, MMRi62 bound to preformed RING domain heterodimers altered the substrate preference toward MDM4 ubiquitination and promoted MDM2-dependent MDM4 degradation in cells. This MDM4-degrader activity of MMRi62 was found to be associated with potent apoptosis induction in leukemia cells. Interestingly, MMRi62 effectively induced apoptosis in p53 mutant, multidrug-resistant leukemia cells and patient samples in addition to p53 wild-type cells. In contrast, MMRi67 as a RING heterodimer disruptor and an enzymatic inhibitor of the MDM2-MDM4 E3 complex lacked MDM4-degrader activity and failed to induce apoptosis in these cells. In summary, this study identifies MMRi62 as a novel MDM2-MDM4-targeting agent and suggests that small molecules capable of promoting MDM4 degradation may be a viable new approach to killing leukemia cells bearing non-functional p53 by apoptosis.
Collapse
Affiliation(s)
- Rati Lama
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Chao Xu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Samuel L. Galster
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Javier Querol-García
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Scott Portwood
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Cory K. Mavis
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Federico M. Ruiz
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Diana Martin
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Jin Wu
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Marianna C. Giorgi
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Jill Bargonetti
- The Department of Biological Sciences, Hunter College, City University of New York, New York, NY, United States
| | - Eunice S. Wang
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | | | - Gerald B. Koudelka
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Sherry R. Chemler
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Inés G. Muñoz
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Xinjiang Wang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- *Correspondence: Xinjiang Wang,
| |
Collapse
|