1
|
Gu K, May HA, Kang MH. Targeting Molecular Signaling Pathways and Cytokine Responses to Modulate c-MYC in Acute Myeloid Leukemia. Front Biosci (Schol Ed) 2024; 16:15. [PMID: 39344393 DOI: 10.31083/j.fbs1603015] [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: 05/21/2024] [Revised: 08/07/2024] [Accepted: 08/18/2024] [Indexed: 10/01/2024]
Abstract
Overexpression of the MYC oncogene, encoding c-MYC protein, contributes to the pathogenesis and drug resistance of acute myeloid leukemia (AML) and many other hematopoietic malignancies. Although standard chemotherapy has predominated in AML therapy over the past five decades, the clinical outcomes and patient response to treatment remain suboptimal. Deeper insight into the molecular basis of this disease should facilitate the development of novel therapeutics targeting specific molecules and pathways that are dysregulated in AML, including fms-like tyrosine kinase 3 (FLT3) gene mutation and cluster of differentiation 33 (CD33) protein expression. Elevated expression of c-MYC is one of the molecular features of AML that determines the clinical prognosis in patients. Increased expression of c-MYC is also one of the cytogenetic characteristics of drug resistance in AML. However, direct targeting of c-MYC has been challenging due to its lack of binding sites for small molecules. In this review, we focused on the mechanisms involving the bromodomain and extra-terminal (BET) and cyclin-dependent kinase 9 (CDK9) proteins, phosphoinositide-Akt-mammalian target of rapamycin (PI3K/AKT/mTOR) and Janus kinase-signal transduction and activation of transcription (JAK/STAT) pathways, as well as various inflammatory cytokines, as an indirect means of regulating MYC overexpression in AML. Furthermore, we highlight Food and Drug Administration (FDA)-approved drugs for AML, and the results of preclinical and clinical studies on novel agents that have been or are currently being tested for efficacy and tolerability in AML therapy. Overall, this review summarizes our current knowledge of the molecular processes that promote leukemogenesis, as well as the various agents that intervene in specific pathways and directly or indirectly modulate c-MYC to disrupt AML pathogenesis and drug resistance.
Collapse
Affiliation(s)
- Kyle Gu
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Harry A May
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Min H Kang
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Pediatrics, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
2
|
Regua AT, Bindal S, Najjar MK, Zhuang C, Khan M, Arrigo ABJ, Gonzalez AO, Zhang XR, Zhu JJ, Watabe K, Lo HW. Dual inhibition of the TrkA and JAK2 pathways using entrectinib and pacritinib suppresses the growth and metastasis of HER2-positive and triple-negative breast cancers. Cancer Lett 2024; 597:217023. [PMID: 38852701 PMCID: PMC11533721 DOI: 10.1016/j.canlet.2024.217023] [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: 04/01/2024] [Revised: 05/26/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
HER2-positive and triple-negative breast cancers (TNBC) are difficult to treat and associated with poor prognosis. Despite showing initial response, HER2-positive breast cancers often acquire resistance to HER2-targeted therapies, and TNBC lack effective therapies. To overcome these clinical challenges, we evaluated the therapeutic utility of co-targeting TrkA and JAK2/STAT3 pathways in these breast cancer subtypes. Here, we report the novel combination of FDA-approved TrkA inhibitors (Entrectinib or Larotrectinib) and JAK2 inhibitors (Pacritinib or Ruxolitinib) synergistically inhibited in vitro growth of HER2-positive breast cancer cells and TNBC cells. The Entrectinib-Pacritinib combination inhibited the breast cancer stem cell subpopulation, reduced expression of stemness genes, SOX2 and MYC, and induced apoptosis. The Entrectinib-Pacritinib combination suppressed orthotopic growth of HER2-positive Trastuzumab-refractory breast cancer xenografts and basal patient-derived xenograft (PDXs), reduced tumoral SOX2 and MYC, and induced apoptosis in both mouse models. The Entrectinib-Pacritinib combination inhibited overall metastatic burden, and brain and bone metastases of intracardially inoculated TNBC cells without toxicity. Together, our results demonstrate for the first time that co-inhibition of TrkA and JAK2 synergistically suppresses breast cancer growth and metastasis, thereby providing preclinical evidence that supports future clinical evaluations.
Collapse
Affiliation(s)
- Angelina T Regua
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shivani Bindal
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mariana K Najjar
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chuling Zhuang
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Munazza Khan
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Austin B J Arrigo
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anneliese O Gonzalez
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xinhai R Zhang
- Department of Pathology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jay-Jiguang Zhu
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Hui-Wen Lo
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
3
|
Zheng X, Chen Z, Guo M, Liang H, Song X, Liu Y, Liao Z, Zhang Y, Guo J, Zhou Y, Zhang ZM, Tu Z, Zhang Y, Chen Y, Zhang Z, Lu X. Structure-Based Optimization of Pyrazinamide-Containing Macrocyclic Derivatives as Fms-like Tyrosine Kinase 3 (FLT3) Inhibitors to Overcome Clinical Mutations. ACS Pharmacol Transl Sci 2024; 7:1485-1506. [PMID: 38751627 PMCID: PMC11092118 DOI: 10.1021/acsptsci.4c00071] [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: 02/08/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 05/18/2024]
Abstract
Secondary mutations in Fms-like tyrosine kinase 3-tyrosine kinase domain (FLT3-TKD) (e.g., D835Y and F691L) have become a major on-target resistance mechanism of FLT3 inhibitors, which present a significant clinical challenge. To date, no effective drugs have been approved to simultaneously overcome clinical resistance caused by these two mutants. Thus, a series of pyrazinamide macrocyclic compounds were first designed and evaluated to overcome the secondary mutations of FLT3. The representative 8v exhibited potent inhibitory activities against FLT3D835Y and FLT3D835Y/F691L with IC50 values of 1.5 and 9.7 nM, respectively. 8v also strongly suppressed the proliferation against Ba/F3 cells transfected with FLT3-ITD, FLT3-ITD-D835Y, FLT3-ITD-F691L, FLT3-ITD-D835Y-F691L, and MV4-11 acute myeloid leukemia (AML) cell lines with IC50 values of 12.2, 10.5, 24.6, 16.9, and 6.8 nM, respectively. Furthermore, 8v demonstrated ideal anticancer efficacy in a Ba/F3-FLT3-ITD-D835Y xenograft model. The results suggested that 8v can serve as a promising macrocycle-based FLT3 inhibitor for the treatment of AML.
Collapse
Affiliation(s)
- Xuan Zheng
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Zhiwen Chen
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Ming Guo
- Department
of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local
Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hong Liang
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Xiaojuan Song
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Yiling Liu
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Zhenling Liao
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Yan Zhang
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Jing Guo
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Yang Zhou
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Zhi-min Zhang
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Zhengchao Tu
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Ye Zhang
- Guangxi
Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Yongheng Chen
- Department
of Oncology, NHC Key Laboratory of Cancer Proteomics, State Local
Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhang Zhang
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
| | - Xiaoyun Lu
- State
Key Laboratory of Bioactive Molecules and Druggability Assessment,
International Cooperative Laboratory of Traditional Chinese Medicine
Modernization and Innovative Drug Discovery of Chinese Ministry of
Education, Guangzhou City Key Laboratory of Precision Chemical Drug
Development, School of Pharmacy, Jinan University, #855 Xingye Avenue, Guangzhou 510632, China
- Department
of Hematology, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou 510632, China
| |
Collapse
|
4
|
Mao F, Gao L, Liu L, Tang Y. Enhanced synergy of pacritinib with temsirolimus and sunitinib in preclinical renal cell carcinoma model by targeting JAK2/STAT pathway. J Chemother 2024; 36:238-248. [PMID: 37916436 DOI: 10.1080/1120009x.2023.2274700] [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: 04/16/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023]
Abstract
Pacritinib is an oral medication that inhibits several kinases including JAK2, FLT3, IRAK and STAT3. It has been recently approved to treat patients with thrombocytopenia and myelofibrosis. Studies are currently exploring the potential use of pacritinib in treating other types of cancer such as leukaemia, breast cancer and prostate cancer. Our study aimed to investigate the effects of pacritinib alone and its combination with standard of care in renal cell carcinoma (RCC). We showed that pacritinib dose-dependently decreased viability of RCC cells, with IC50 at nanomolar or low micromolar concentration rage. Pacritinib inhibited cell proliferation, decreased colony formation, and increased apoptosis. Interestingly, pacritinib exhibited synergistic effects when combined with temsirolimus and sunitinib, but antagonistic effects when combined with doxorubicin, in a panel of RCC cell lines. We also confirmed that the combination of pacritinib with temsirolimus and sunitinib resulted in synergistic effects in RCC mouse models, with complete inhibition of tumour growth throughout the treatment period. Mechanistic studies indicated that the inhibition of JAK2, but not IRAK, was the main contributor to the anti-RCC activity of pacritinib. Our study is the first to demonstrate that pacritinib shows promise as a treatment option for RCC and underscores the therapeutic potential of targeting the JAK2/STAT signalling pathway in RCC.
Collapse
Affiliation(s)
- Fei Mao
- Department of Urology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, People's Republic of China
| | - Liangkui Gao
- Department of Urology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, People's Republic of China
| | - Liming Liu
- Department of Urology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, People's Republic of China
| | - Yuanjia Tang
- Department of Urology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, People's Republic of China
| |
Collapse
|
5
|
Seipel K, Mandhair H, Bacher U, Pabst T. FLT3 and IRAK4 Inhibitor Emavusertib in Combination with BH3-Mimetics in the Treatment of Acute Myeloid Leukemia. Curr Issues Mol Biol 2024; 46:2946-2960. [PMID: 38666914 PMCID: PMC11049208 DOI: 10.3390/cimb46040184] [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: 02/27/2024] [Revised: 03/22/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Targeting the FLT3 receptor and the IL-1R associated kinase 4 as well as the anti-apoptotic proteins MCL1 and BCL2 may be a promising novel approach in the treatment of acute myeloid leukemia (AML). The FLT3 and IRAK4 inhibitor emavusertib (CA4948), the MCL1 inhibitor S63845, the BCL2 inhibitor venetoclax, and the HSP90 inhibitor PU-H71 were assessed as single agents and in combination for their ability to induce apoptosis and cell death in leukemic cells in vitro. AML cells represented all major morphologic and molecular subtypes, including FLT3-ITD and NPM1 mutant AML cell lines and a variety of patient-derived AML cells. Emavusertib in combination with MCL1 inhibitor S63845 or BCL2 inhibitor venetoclax induced cell cycle arrest and apoptosis in MOLM-13 cells. In primary AML cells, the response to emavusertib was associated with the presence of the FLT3 gene mutation with an allelic ratio >0.5 and the presence of NPM1 gene mutations. S63845 was effective in all tested AML cell lines and primary AML samples. Blast cell percentage was positively associated with the response to CA4948, S63845, and venetoclax, with elevated susceptibility of primary AML with blast cell fraction >80%. Biomarkers of the response to venetoclax included the blast cell percentage and bone marrow infiltration rate, as well as the expression levels of CD11b, CD64, and CD117. Elevated susceptibility to CA4948 combination treatments with S63845 or PU-H71 was associated with FLT3-mutated AML and CD34 < 30%. The combination of CA4948 and BH3-mimetics may be effective in the treatment in FLT3-mutated AML with differential target specificity for MCL1 and BCL2 inhibitors. Moreover, the combination of CA4948 and PU-H71 may be a candidate combination treatment in FLT3-mutated AML.
Collapse
Affiliation(s)
- Katja Seipel
- Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland;
| | - Harpreet Mandhair
- Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland;
| | - Ulrike Bacher
- Department of Hematology, University Hospital Bern, 3010 Bern, Switzerland;
| | - Thomas Pabst
- Department of Medical Oncology, University Hospital Bern, 3010 Bern, Switzerland
| |
Collapse
|
6
|
Feng D, Liu B, Chen Z, Xu J, Geng M, Duan W, Ai J, Zhang H. Discovery of hematopoietic progenitor kinase 1 inhibitors using machine learning-based screening and free energy perturbation. J Biomol Struct Dyn 2024:1-13. [PMID: 38198294 DOI: 10.1080/07391102.2024.2301754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is a key negative regulator of T-cell receptor (TCR) signaling and a promising target for cancer immunotherapy. The development of novel HPK1 inhibitors is challenging yet promising. In this study, we used a combination of machine learning (ML)-based virtual screening and free energy perturbation (FEP) calculations to identify novel HPK1 inhibitors. ML-based screening yielded 10 potent HPK1 inhibitors (IC50 < 1 μM). The FEP-guided modification of the in-house false-positive hit, DW21302, revealed that a single key atom change could trigger activity cliffs. The resulting DW21302-A was a potent HPK1 inhibitor (IC50 = 2.1 nM) and potently inhibited cellular HPK1 signaling and enhanced T-cell function. Molecular dynamics (MD) simulations and ADME predictions confirmed DW21302-A as candidate compound. This study provides new strategies and chemical scaffolds for HPK1 inhibitor development.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Dazhi Feng
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Bo Liu
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| | - Zhiwei Chen
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Meiyu Geng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Wenhu Duan
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Jing Ai
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hefeng Zhang
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
7
|
Wei XH, Liu YY. Potential applications of JAK inhibitors, clinically approved drugs against autoimmune diseases, in cancer therapy. Front Pharmacol 2024; 14:1326281. [PMID: 38235120 PMCID: PMC10792058 DOI: 10.3389/fphar.2023.1326281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024] Open
Abstract
Disturbances in immunoregulation may lead to both cancer and autoimmune diseases. Many therapeutic drugs for autoimmune diseases also display anti-tumor efficacy. The Janus kinase/signal transducer and activator of transcription signaling pathways are involved in the secretion of more than 50 distinct cytokines, which have critical roles in inducing autoimmune diseases and tumorigenesis. Thus, Janus kinases have become classical immunotherapeutic targets for immune disease. More than 70 Janus kinase inhibitors have been approved as immunomodulatory drugs for clinical use, of which 12 are used in the treatment of autoimmune diseases. This systematic review aims to elucidate the anti-tumor role of clinically approved Janus kinase inhibitors that were primarily designed for the treatment of autoimmune diseases and their potential for clinical translation as cancer treatments.
Collapse
Affiliation(s)
- Xiao-Huan Wei
- Respiratory and Critical Care Department, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Oncology Department, People’s Hospital of Peixian, Xuzhou, Jiangsu, China
| | - Yuan-Yuan Liu
- Respiratory and Critical Care Department, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
8
|
Putnam CM, Kondeti L, Kesler MBA, Varney ME. Modulating the immune system as a therapeutic target for myelodysplastic syndromes and acute myeloid leukemia. Biochem Cell Biol 2023; 101:481-495. [PMID: 37566901 DOI: 10.1139/bcb-2022-0374] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2023] Open
Abstract
Modulating the immune system to treat diseases, including myeloid malignancies, has resulted in the development of a multitude of novel therapeutics in recent years. Myelodysplastic syndromes or neoplasms (MDS) and acute myeloid leukemia (AML) are hematologic malignancies that arise from defects in hematopoietic stem and progenitor cells (HSPCs). Dysregulated immune responses, especially in innate immune and inflammatory pathways, are highly associated with the acquisition of HSPC defects in MDS and AML pathogenesis. In addition to utilizing the immune system in immunotherapeutic interventions such as chimeric antigen receptor T cell therapy, vaccines, and immune checkpoint inhibitors, mitigating dysregulation of innate immune and inflammatory responses in MDS and AML remains a priority in slowing the initiation and progression of these myeloid malignancies. This review provides a comprehensive summary of the current progress of diverse strategies to utilize or modulate the immune system in the treatment of MDS and AML.
Collapse
Affiliation(s)
- Caroline M Putnam
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV, USA
| | - Lahari Kondeti
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV, USA
| | - Meredith B A Kesler
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV, USA
| | - Melinda E Varney
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV, USA
| |
Collapse
|
9
|
Swaminathan M, Aly MM, Khan AM, Share BA, Dhillon V, Lalo E, Ramos H, Akers KG, Kim S, Balasubramanian S. Efficacy analysis of different FLT3 inhibitors in patients with relapsed/refractory acute myeloid leukemia and high-risk myelodysplastic syndrome. EJHAEM 2023; 4:165-173. [PMID: 36819163 PMCID: PMC9928788 DOI: 10.1002/jha2.616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022]
Abstract
Several FLT3 inhibitors(i) are available to treat relapsed/refractory (R/R) FLT3-internal tandem duplicated acute myeloid leukemia (AML). This study analyzes the efficacies of various FLT3i (types 1 and 2) tested in clinical trials in treating R/R AML and high-risk myelodysplastic syndromes (HR-MDS). PubMed and EMBASE databases were searched for single/double-arm phase I/II/III R/R AML or HR-MDS clinical trials published between 1/1/2000 and 6/1/2021. The outcomes studied were composite response rate (CRc) and overall response rate (ORR). Toxicities were compared based on the organ system. The 28 studies analyzed had 1927 patients. The pooled ORR and (CRc) for all FLT3i were 53% (95% CI, 43%-63%) and 34% (95% CI, 26%-44%). Pooled ORR and CRc were 37% (95% CI, 25%-51%) and 35% (95% CI, 21%-52%) for type 1 and 58% (95% CI, 43%-71%) and 38% (95% CI, 27%-50%) for type 2, respectively. Gastrointestinal (GI) and hematological toxicity occurred in 22% (95% CI, 19%-25.4%) and 74.6% (95% CI, 70%-79%) with type 1 and 13.9% (95% CI, 12%-16%) and 57.7% (95% CI, 54.6%-60.8%) with type 2 FLT3i. QTc prolongation occurred in 2.06% (95% CI, 1.03%-3.65%) with type 1 and 7% (95% CI, 5.3%-9%) with type 2 FLT3i. Type 2 FLT3i had less GI toxicity but more QTc prolongation. Prospective studies are needed to compare the efficacy of type 1 and 2 FLT3i.
Collapse
Affiliation(s)
- Mahesh Swaminathan
- Department of MedicineRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Mai M. Aly
- Clinical Hematology UnitInternal Medicine DepartmentAssiut University HospitalAssiutEgypt
| | - Abdul Moiz Khan
- Department of OncologyKarmanos Cancer InstituteWayne State UniversityDetroitMichiganUSA
| | - Bayan Al Share
- Department of OncologyKarmanos Cancer InstituteWayne State UniversityDetroitMichiganUSA
| | - Vikram Dhillon
- Department of Internal MedicineWayne State University School of MedicineDetroitMichiganUSA
| | - Enxhi Lalo
- Wayne State University School of MedicineDetroitMichiganUSA
| | - Harry Ramos
- Wayne State University School of MedicineDetroitMichiganUSA
| | | | - Seongho Kim
- Department of OncologyKarmanos Cancer InstituteWayne State UniversityDetroitMichiganUSA
- Biostatistics and Bioinformatics CoreKarmanos Cancer InstituteWayne State UniversityDetroitMichiganUSA
| | - Suresh Balasubramanian
- Department of OncologyKarmanos Cancer InstituteWayne State UniversityDetroitMichiganUSA
- Translational Hematology and Oncology ResearchTaussig Cancer Institute, Cleveland ClinicClevelandOhioUSA
| |
Collapse
|
10
|
Ghezzi C, Chen BY, Damoiseaux R, Clark PM. Pacritinib inhibits glucose consumption in squamous cell lung cancer cells by targeting FLT3. Sci Rep 2023; 13:1442. [PMID: 36697489 PMCID: PMC9876922 DOI: 10.1038/s41598-023-28576-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Squamous cell lung cancer maintains its growth through elevated glucose consumption, but selective glucose consumption inhibitors are lacking. Here, we discovered using a high-throughput screen new compounds that block glucose consumption in three squamous cell lung cancer cell lines and identified 79 compounds that block glucose consumption in one or more of these cell lines. Based on its ability to block glucose consumption in all three cell lines, pacritinib, an inhibitor of FMS Related Receptor Tyrosine Kinase 3 (FLT3) and Janus Kinase 2 (JAK2), was further studied. Pacritinib decreased glucose consumption in squamous cell lung cancer cells in cell culture and in vivo without affecting glucose consumption in healthy tissues. Pacritinib blocked hexokinase activity, and Hexokinase 1 and 2 mRNA and protein expression. Overexpression of Hexokinase 1 blocked the ability of pacritinib to inhibit glucose consumption in squamous cell lung cancer cells. Overexpression of FLT3 but not JAK2 significantly increased glucose consumption and blocked the ability of pacritinib to inhibit glucose consumption in squamous cell lung cancer cells. Additional FLT3 inhibitors blocked glucose consumption in squamous cell lung cancer cells. Our study identifies FLT3 inhibitors as a new class of inhibitors that can block glucose consumption in squamous cell lung cancer.
Collapse
Affiliation(s)
- Chiara Ghezzi
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Bao Ying Chen
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Robert Damoiseaux
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Peter M Clark
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA.
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
11
|
Tecik M, Adan A. Therapeutic Targeting of FLT3 in Acute Myeloid Leukemia: Current Status and Novel Approaches. Onco Targets Ther 2022; 15:1449-1478. [PMID: 36474506 PMCID: PMC9719701 DOI: 10.2147/ott.s384293] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/19/2022] [Indexed: 08/13/2023] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) is mutated in approximately 30% of acute myeloid leukemia (AML) patients. The presence of FLT3-ITD (internal tandem duplication, 20-25%) mutation and, to a lesser extent, FLT3-TKD (tyrosine kinase domain, 5-10%) mutation is associated with poorer diagnosis and therapy response since the leukemic cells become hyperproliferative and resistant to apoptosis after continuous activation of FLT3 signaling. Targeting FLT3 has been the focus of many pre-clinical and clinical studies. Hence, many small-molecule FLT3 inhibitors (FLT3is) have been developed, some of which are approved such as midostaurin and gilteritinib to be used in different clinical settings, either in combination with chemotherapy or alone. However, many questions regarding the best treatment strategy remain to be answered. On the other hand, various FLT3-dependent and -independent resistance mechanisms could be evolved during FLT3i therapy which limit their clinical impact. Therefore, identifying molecular mechanisms of resistance and developing novel strategies to overcome this obstacle is a current interest in the field. In this review, recent studies of approved FLT3i and knowledge about major resistance mechanisms of clinically approved FLT3i's will be discussed together with novel treatment approaches such as designing novel FLT3i and dual FLT3i and combination strategies including approved FLT3i plus small-molecule agents targeting altered molecules in the resistant cells to abrogate resistance. Moreover, how to choose an appropriate FLT3i for the patients will be summarized based on what is currently known from available clinical data. In addition, strategies beyond FLT3i's including immunotherapeutics, small-molecule FLT3 degraders, and flavonoids will be summarized to highlight potential alternatives in FLT3-mutated AML therapy.
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
|
12
|
Ge SS, Liu SB, Xue SL. Developments and challenges of FLT3 inhibitors in acute myeloid leukemia. Front Oncol 2022; 12:996438. [PMID: 36185253 PMCID: PMC9515417 DOI: 10.3389/fonc.2022.996438] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
FLT3 mutations are one of the most common genetic alterations in acute myeloid leukemia (AML) and are identified in approximately one-third of newly diagnosed patients. Aberrant FLT3 receptor signaling has important implications for the biology and clinical management of AML. In recent years, targeting FLT3 has been a part of every course of treatment in FLT3-ITD/TKD-mutated AML and contributes to substantially prolonged survival. At the same time, wide application of next-generation sequencing (NGS) technology has revealed a series of non-canonical FLT3 mutations, including point mutations and small insertions/deletions. Some of these mutations may be able to influence downstream phosphorylation and sensitivity to FLT3 inhibitors, while the correlation with clinical outcomes remains unclear. Exploration of FLT3-targeted therapy has made substantial progress, but resistance to FLT3 inhibitors has become a pressing issue. The mechanisms underlying FLT3 inhibitor tolerance can be roughly divided into primary resistance and secondary resistance. Primary resistance is related to abnormalities in signaling factors, such as FL, CXCL12, and FGF2, and secondary resistance mainly involves on-target mutations and off-target aberrations. To overcome this problem, novel agents such as FF-10101 have shown promising potential. Multitarget strategies directed at FLT3 and anomalous signaling factors simultaneously are in active clinical development and show promising results.
Collapse
Affiliation(s)
- Shuai-Shuai Ge
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Song-Bai Liu
- Suzhou Key Laboratory of Medical Biotechnology, Suzhou Vocational Health College, Suzhou, China
| | - Sheng-Li Xue
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
13
|
Overcoming Resistance: FLT3 Inhibitors Past, Present, Future and the Challenge of Cure. Cancers (Basel) 2022; 14:cancers14174315. [PMID: 36077850 PMCID: PMC9454516 DOI: 10.3390/cancers14174315] [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: 05/26/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
FLT3 ITD and TKD mutations occur in 20% and 10% of Acute Myeloid Leukemia (AML), respectively, and they represent the target of the first approved anti-leukemic therapies in the 2000s. Type I and type II FLT3 inhibitors (FLT3i) are active against FLT3 TKD/ITD and FLT3 ITD mutations alone respectively, but they still fail remissions in 30-40% of patients due to primary and secondary mechanisms of resistance, with variable relapse rate of 30-50%, influenced by NPM status and FLT3 allelic ratio. Mechanisms of resistance to FLT3i have recently been analyzed through NGS and single cell assays that have identified and elucidated the polyclonal nature of relapse in clinical and preclinical studies, summarized here. Knowledge of tumor escape pathways has helped in the identification of new targeted drugs to overcome resistance. Immunotherapy and combination or sequential use of BCL2 inhibitors and experimental drugs including aurora kinases, menin and JAK2 inhibitors will be the goal of present and future clinical trials, especially in patients with FLT3-mutated (FLT3mut) AML who are not eligible for allogeneic transplantation.
Collapse
|
14
|
Gerds AT, Gotlib J, Ali H, Bose P, Dunbar A, Elshoury A, George TI, Gundabolu K, Hexner E, Hobbs GS, Jain T, Jamieson C, Kaesberg PR, Kuykendall AT, Madanat Y, McMahon B, Mohan SR, Nadiminti KV, Oh S, Pardanani A, Podoltsev N, Rein L, Salit R, Stein BL, Talpaz M, Vachhani P, Wadleigh M, Wall S, Ward DC, Bergman MA, Hochstetler C. Myeloproliferative Neoplasms, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2022; 20:1033-1062. [PMID: 36075392 DOI: 10.6004/jnccn.2022.0046] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The classic Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) consist of myelofibrosis, polycythemia vera, and essential thrombocythemia and are a heterogeneous group of clonal blood disorders characterized by an overproduction of blood cells. The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for MPN were developed as a result of meetings convened by a multidisciplinary panel with expertise in MPN, with the goal of providing recommendations for the management of MPN in adults. The Guidelines include recommendations for the diagnostic workup, risk stratification, treatment, and supportive care strategies for the management of myelofibrosis, polycythemia vera, and essential thrombocythemia. Assessment of symptoms at baseline and monitoring of symptom status during the course of treatment is recommended for all patients. This article focuses on the recommendations as outlined in the NCCN Guidelines for the diagnosis of MPN and the risk stratification, management, and supportive care relevant to MF.
Collapse
Affiliation(s)
- Aaron T Gerds
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | - Haris Ali
- City of Hope National Medical Center
| | | | | | | | | | | | | | | | - Tania Jain
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | | | | | | | - Stephen Oh
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | - Rachel Salit
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | - Brady L Stein
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | - Sarah Wall
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Dawn C Ward
- UCLA Jonsson Comprehensive Cancer Center; and
| | | | | |
Collapse
|
15
|
Acharya B, Saha D, Armstrong D, Lakkaniga NR, Frett B. FLT3 inhibitors for acute myeloid leukemia: successes, defeats, and emerging paradigms. RSC Med Chem 2022; 13:798-816. [PMID: 35923716 PMCID: PMC9298189 DOI: 10.1039/d2md00067a] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/21/2022] [Indexed: 09/10/2023] Open
Abstract
FLT3 mutations are one of the most common genetic aberrations found in nearly 30% of acute myeloid leukemias (AML). The mutations are associated with poor prognosis despite advances in the understanding of the biological mechanisms of AML. Numerous small molecule FLT3 inhibitors have been developed in an effort to combat AML. Even with the development of these inhibitors, the five-year overall survival for newly diagnosed AML is less than 30%. In 2017, midostaurin received FDA approval to treat AML, which was the first approved FLT3 inhibitor in the U.S. and Europe. Following, gilteritinib received FDA approval in 2018 and in 2019 quizartinib received approval in Japan. This review parallels these clinical success stories along with other pre-clinical and clinical investigations of FLT3 inhibitors.
Collapse
Affiliation(s)
- Baku Acharya
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Debasmita Saha
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Daniel Armstrong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| | - Naga Rajiv Lakkaniga
- Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines) Dhanbad Jharkhand 826004 India
| | - Brendan Frett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences Little Rock AR 72205 USA
| |
Collapse
|
16
|
Desikan SP, Daver N, DiNardo C, Kadia T, Konopleva M, Ravandi F. Resistance to targeted therapies: delving into FLT3 and IDH. Blood Cancer J 2022; 12:91. [PMID: 35680852 PMCID: PMC9184476 DOI: 10.1038/s41408-022-00687-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Recent advances in FLT3 and IDH targeted inhibition have improved response rates and overall survival in patients with mutations affecting these respective proteins. Despite this success, resistance mechanisms have arisen including mutations that disrupt inhibitor-target interaction, mutations impacting alternate pathways, and changes in the microenvironment. Here we review the role of these proteins in leukemogenesis, their respective inhibitors, mechanisms of resistance, and briefly ongoing studies aimed at overcoming resistance.
Collapse
Affiliation(s)
- Sai Prasad Desikan
- Departments of Leukemia, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Houston, TX, USA
| | - Naval Daver
- Departments of Leukemia, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Houston, TX, USA
| | - Courtney DiNardo
- Departments of Leukemia, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Houston, TX, USA
| | - Tapan Kadia
- Departments of Leukemia, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Houston, TX, USA
| | - Marina Konopleva
- Departments of Leukemia, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Houston, TX, USA
| | - Farhad Ravandi
- Departments of Leukemia, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Houston, TX, USA.
| |
Collapse
|
17
|
A Comprehensive Overview of Globally Approved JAK Inhibitors. Pharmaceutics 2022; 14:pharmaceutics14051001. [PMID: 35631587 PMCID: PMC9146299 DOI: 10.3390/pharmaceutics14051001] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
Janus kinase (JAK) is a family of cytoplasmic non-receptor tyrosine kinases that includes four members, namely JAK1, JAK2, JAK3, and TYK2. The JAKs transduce cytokine signaling through the JAK-STAT pathway, which regulates the transcription of several genes involved in inflammatory, immune, and cancer conditions. Targeting the JAK family kinases with small-molecule inhibitors has proved to be effective in the treatment of different types of diseases. In the current review, eleven of the JAK inhibitors that received approval for clinical use have been discussed. These drugs are abrocitinib, baricitinib, delgocitinib, fedratinib, filgotinib, oclacitinib, pacritinib, peficitinib, ruxolitinib, tofacitinib, and upadacitinib. The aim of the current review was to provide an integrated overview of the chemical and pharmacological data of the globally approved JAK inhibitors. The synthetic routes of the eleven drugs were described. In addition, their inhibitory activities against different kinases and their pharmacological uses have also been explained. Moreover, their crystal structures with different kinases were summarized, with a primary focus on their binding modes and interactions. The proposed metabolic pathways and metabolites of these drugs were also illustrated. To sum up, the data in the current review could help in the design of new JAK inhibitors with potential therapeutic benefits in inflammatory and autoimmune diseases.
Collapse
|
18
|
Expanding the structure-activity relationship of cytotoxic diphenyl macrocycles. Bioorg Med Chem Lett 2022; 62:128628. [PMID: 35182774 DOI: 10.1016/j.bmcl.2022.128628] [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: 11/28/2021] [Revised: 02/08/2022] [Accepted: 02/13/2022] [Indexed: 11/21/2022]
Abstract
Twenty-four biaryl tetrapeptide macrocycles were synthesized as an extension of our previous work. Two groups of compounds were constructed for establishing a structure-activity relationship: one having an aromatic substituent at α-position of one exo-peptide and the other group with a variation in the size of the lipophilic chain. Compound 13t had the best cytotoxicity from all the compounds tested (in a panel of six human cancer cell lines) and low toxicity on one healthy cell line. The study identified the lipophilic chain as the main structural moiety for improving the biological activity, being the seven-carbon chain the optimal length. On the other hand, the aromatic rings at α-position did not enhance the cytotoxicity.
Collapse
|
19
|
Solana-Altabella A, Ballesta-López O, Megías-Vericat JE, Martínez-Cuadrón D, Montesinos P. Emerging FLT3 inhibitors for the treatment of acute myeloid leukemia. Expert Opin Emerg Drugs 2022; 27:1-18. [DOI: 10.1080/14728214.2021.2009800] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Antonio Solana-Altabella
- Servicio de Farmacia, Área del Medicamento. Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia– Spain
- Instituto de Investigación Sanitaria La Fe (IISLAFE). Av. Fernando Abril Martorell, Valencia–Spain
| | - Octavio Ballesta-López
- Servicio de Farmacia, Área del Medicamento. Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia– Spain
- Instituto de Investigación Sanitaria La Fe (IISLAFE). Av. Fernando Abril Martorell, Valencia–Spain
| | - Juan Eduardo Megías-Vericat
- Servicio de Farmacia, Área del Medicamento. Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia– Spain
| | - David Martínez-Cuadrón
- Servicio de Farmacia, Área del Medicamento. Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia– Spain
- Instituto de Investigación Sanitaria La Fe (IISLAFE). Av. Fernando Abril Martorell, Valencia–Spain
| | - Pau Montesinos
- Instituto de Investigación Sanitaria La Fe (IISLAFE). Av. Fernando Abril Martorell, Valencia–Spain
- Servicio de Hematología y Hemoterapia. Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia-Spain
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Cell intrinsic and extrinsic perturbations to inflammatory signaling pathways are a hallmark of development and progression of hematologic malignancies. The interleukin 1 receptor-associated kinases (IRAKs) are a family of related signaling intermediates (IRAK1, IRAK2, IRAK3, IRAK4) that operate at the nexus of multiple inflammatory pathways implicated in the hematologic malignancies. In this review, we explicate the oncogenic role of these kinases and review recent therapeutic advances in the dawning era of IRAK-targeted therapy. RECENT FINDINGS Emerging evidence places IRAK signaling at the confluence of adaptive resistance and oncogenesis in the hematologic malignancies and solid tissue tumors. Preclinical investigations nominate the IRAK kinases as targetable molecular dependencies in diverse cancers. SUMMARY IRAK-targeted therapies that have matriculated to early phase trials are yielding promising preliminary results. However, studies of IRAK kinase signaling continue to defy conventional signaling models and raise questions as to the design of optimal treatment strategies. Efforts to refine IRAK signaling mechanisms in the malignant context will inspire deliberate IRAK-targeted drug development and informed combination therapy.
Collapse
Affiliation(s)
- Joshua Bennett
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center
- Department of Cancer Biology
| | - Daniel T. Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center
- Department of Cancer Biology
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
21
|
IRAK1-regulated IFN-γ signaling induces MDSC to facilitate immune evasion in FGFR1-driven hematological malignancies. Mol Cancer 2021; 20:165. [PMID: 34906138 PMCID: PMC8670266 DOI: 10.1186/s12943-021-01460-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 11/16/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Stem Cell leukemia/lymphoma syndrome (SCLL) presents as a myeloproliferative disease which can progress to acute myeloid leukemia and is associated with the coincident development of B-cell and T-cell lymphomas. SCLL is driven by the constitutive activation of fibroblast growth factor receptor-1 (FGFR1) as a result of chromosome translocations with poor outcome. Mouse models have been developed which faithfully recapitulate the human disease and have been used to characterize the molecular genetic events that are associated with development and progression of the disease. METHODS CRISPR/Cas9 approaches were used to generate SCLL cells null for Interleukin receptor associated kinase 1 (IRAK1) and interferon gamma (IFNG) which were introduced into syngeneic hosts through tail vein injection. Development of the disease and changes in immune cell composition and activity were monitored using flow cytometry. Bead-based immunoassays were used to compare the cytokine and chemokine profiles of control and knock out (KO) cells. Antibody mediated, targeted depletion of T cell and MDSCs were performed to evaluate their role in antitumor immune responses. RESULTS In SCLL, FGFR1 activation silences miR-146b-5p through DNMT1-mediated promoter methylation, which derepresses the downstream target IRAK1. IRAK1 KO SCLL cells were xenografted into immunocompetent syngeneic mice where the typical rapid progression of disease was lost and the mice remained disease free. IRAK1 in this system has no effect on cell cycle progression or apoptosis and robust growth of the KO cells in immunodeficient mice suggested an effect on immune surveillance. Depletion of T-cells in immunocompetent mice restored leukemogenesis of the KO cells, and tumor killing assays confirmed the role of T cells in tumor clearance. Analysis of the immune cell profile in mice transplanted with the IRAK1 expressing mock control (MC) cells shows that there is an increase in levels of myeloid-derived suppressor cells (MDSCs) with a concomitant decrease in CD4+/CD8+ T-cell levels. MDSC suppression assays and depletion experiments showed that these MDSCs were responsible for suppression of the T cell mediated leukemia cell elimination. Immuno-profiling of a panel of secreted cytokines and chemokines showed that activation of IFN-γ is specifically impaired in the KO cells. In vitro and in vivo expression assays and engraftment with interferon gamma receptor-1 (IFNGR1) null mice and IFNG KO SCLL cells, showed the leukemia cells produced IFN-γ directly participating in the induction of MDSCs to establish immune evasion. Inhibition of IRAK1 using pacritinib suppresses leukemogenesis with impaired induction of MDSCs and attenuated suppression of CD4+/CD8+ T-cells. CONCLUSIONS IRAK1 orchestrates a previously unknown FGFR1-directed immune escape mechanism in SCLL, through induction of MDSCs via regulation of IFN-γ signaling from leukemia cells, and targeting IRAK1 may provide a means of suppressing tumor growth in this syndrome by restoring immune surveillance.
Collapse
|
22
|
Second-Generation Jak2 Inhibitors for Advanced Prostate Cancer: Are We Ready for Clinical Development? Cancers (Basel) 2021; 13:cancers13205204. [PMID: 34680353 PMCID: PMC8533841 DOI: 10.3390/cancers13205204] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/05/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Prostate Cancer (PC) is currently estimated to affect 1 in 9 men and is the second leading cause of cancer in men in the US. While androgen deprivation therapy, which targets the androgen receptor, is one of the front-line therapies for advanced PC and for recurrence of organ-confined PC treated with surgery, lethal castrate-resistant PC develops consistently in patients. PC is a multi-focal cancer with different grade carcinoma areas presenting simultaneously. Jak2-Stat5 signaling pathway has emerged as a potentially highly effective molecular target in PCs with positive areas for activated Stat5 protein. Activated Jak2-Stat5 signaling can be readily targeted by the second-generation Jak2-inhibitors that have been developed for myeloproliferative and autoimmune disorders and hematological malignancies. In this review, we analyze and summarize the Jak2 inhibitors that are currently in preclinical and clinical development. Abstract Androgen deprivation therapy (ADT) for metastatic and high-risk prostate cancer (PC) inhibits growth pathways driven by the androgen receptor (AR). Over time, ADT leads to the emergence of lethal castrate-resistant PC (CRPC), which is consistently caused by an acquired ability of tumors to re-activate AR. This has led to the development of second-generation anti-androgens that more effectively antagonize AR, such as enzalutamide (ENZ). However, the resistance of CRPC to ENZ develops rapidly. Studies utilizing preclinical models of PC have established that inhibition of the Jak2-Stat5 signaling leads to extensive PC cell apoptosis and decreased tumor growth. In large clinical cohorts, Jak2-Stat5 activity predicts PC progression and recurrence. Recently, Jak2-Stat5 signaling was demonstrated to induce ENZ-resistant PC growth in preclinical PC models, further emphasizing the importance of Jak2-Stat5 for therapeutic targeting for advanced PC. The discovery of the Jak2V617F somatic mutation in myeloproliferative disorders triggered the rapid development of Jak1/2-specific inhibitors for a variety of myeloproliferative and auto-immune disorders as well as hematological malignancies. Here, we review Jak2 inhibitors targeting the mutated Jak2V617F vs. wild type (WT)-Jak2 that are currently in the development pipeline. Among these 35 compounds with documented Jak2 inhibitory activity, those with potency against WT-Jak2 hold strong potential for advanced PC therapy.
Collapse
|
23
|
Moser B, Edtmayer S, Witalisz-Siepracka A, Stoiber D. The Ups and Downs of STAT Inhibition in Acute Myeloid Leukemia. Biomedicines 2021; 9:1051. [PMID: 34440253 PMCID: PMC8392322 DOI: 10.3390/biomedicines9081051] [Citation(s) in RCA: 9] [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: 06/30/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 01/03/2023] Open
Abstract
Aberrant Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling is implicated in the pathogenesis of acute myeloid leukemia (AML), a highly heterogeneous hematopoietic malignancy. The management of AML is complex and despite impressive efforts into better understanding its underlying molecular mechanisms, survival rates in the elderly have not shown a substantial improvement over the past decades. This is particularly due to the heterogeneity of AML and the need for personalized approaches. Due to the crucial role of the deregulated JAK-STAT signaling in AML, selective targeting of the JAK-STAT pathway, particularly constitutively activated STAT3 and STAT5 and their associated upstream JAKs, is of great interest. This strategy has shown promising results in vitro and in vivo with several compounds having reached clinical trials. Here, we summarize recent FDA approvals and current potential clinically relevant inhibitors for AML patients targeting JAK and STAT proteins. This review underlines the need for detailed cytogenetic analysis and additional assessment of JAK-STAT pathway activation. It highlights the ongoing development of new JAK-STAT inhibitors with better disease specificity, which opens up new avenues for improved disease management.
Collapse
Affiliation(s)
| | | | | | - Dagmar Stoiber
- Department of Pharmacology, Physiology and Microbiology, Division Pharmacology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (B.M.); (S.E.); (A.W.-S.)
| |
Collapse
|
24
|
Zhan D, Park CY. Stem Cells in the Myelodysplastic Syndromes. FRONTIERS IN AGING 2021; 2:719010. [PMID: 35822030 PMCID: PMC9261372 DOI: 10.3389/fragi.2021.719010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/02/2021] [Indexed: 01/12/2023]
Abstract
The myelodysplastic syndromes (MDS) represent a group of clonal disorders characterized by ineffective hematopoiesis, resulting in peripheral cytopenias and frequent transformation to acute myeloid leukemia (AML). We and others have demonstrated that MDS arises in, and is propagated by malignant stem cells (MDS-SCs), that arise due to the sequential acquisition of genetic and epigenetic alterations in normal hematopoietic stem cells (HSCs). This review focuses on recent advancements in the cellular and molecular characterization of MDS-SCs, as well as their role in mediating MDS clinical outcomes. In addition to discussing the cell surface proteins aberrantly upregulated on MDS-SCs that have allowed the identification and prospective isolation of MDS-SCs, we will discuss the recurrent cytogenetic abnormalities and genetic mutations present in MDS-SCs and their roles in initiating disease, including recent studies demonstrating patterns of clonal evolution and disease progression from pre-malignant HSCs to MDS-SCs. We also will discuss the pathways that have been described as drivers or promoters of disease, including hyperactivated innate immune signaling, and how the identification of these alterations in MDS-SC have led to investigations of novel therapeutic strategies to treat MDS. It is important to note that despite our increasing understanding of the pathogenesis of MDS, the molecular mechanisms that drive responses to therapy remain poorly understood, especially the mechanisms that underlie and distinguish hematologic improvement from reductions in blast burden. Ultimately, such distinctions will be required in order to determine the shared and/or unique molecular mechanisms that drive ineffective hematopoiesis, MDS-SC maintenance, and leukemic transformation.
Collapse
Affiliation(s)
- Di Zhan
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, United States
| | - Christopher Y. Park
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, United States
- *Correspondence: Christopher Y. Park,
| |
Collapse
|
25
|
Wang Z, Cai J, Cheng J, Yang W, Zhu Y, Li H, Lu T, Chen Y, Lu S. FLT3 Inhibitors in Acute Myeloid Leukemia: Challenges and Recent Developments in Overcoming Resistance. J Med Chem 2021; 64:2878-2900. [PMID: 33719439 DOI: 10.1021/acs.jmedchem.0c01851] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the FMS-like tyrosine kinase 3 (FLT3) gene are often present in newly diagnosed acute myeloid leukemia (AML) patients with an incidence rate of approximately 30%. Recently, many FLT3 inhibitors have been developed and exhibit positive preclinical and clinical effects against AML. However, patients develop resistance soon after undergoing FLT3 inhibitor treatment, resulting in short durable responses and poor clinical effects. This review will discuss the main mechanisms of resistance to clinical FLT3 inhibitors and summarize the emerging strategies that are utilized to overcome drug resistance. Basically, medicinal chemistry efforts to develop new small-molecule FLT3 inhibitors offer a direct solution to this problem. Other potential strategies include the combination of FLT3 inhibitors with other therapies and the development of multitarget inhibitors. It is hoped that this review will provide inspiring insights into the discovery of new AML therapies that can eventually overcome the resistance to current FLT3 inhibitors.
Collapse
Affiliation(s)
- Zhijie Wang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jiongheng Cai
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jie Cheng
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Wenqianzi Yang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Yifan Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Hongmei Li
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P.R. China
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| |
Collapse
|
26
|
Kennedy VE, Smith CC. FLT3 Mutations in Acute Myeloid Leukemia: Key Concepts and Emerging Controversies. Front Oncol 2021; 10:612880. [PMID: 33425766 PMCID: PMC7787101 DOI: 10.3389/fonc.2020.612880] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/19/2020] [Indexed: 12/27/2022] Open
Abstract
The FLT3 receptor is overexpressed on the majority of acute myeloid leukemia (AML) blasts. Mutations in FLT3 are the most common genetic alteration in AML, identified in approximately one third of newly diagnosed patients. FLT3 internal tandem duplication mutations (FLT3-ITD) are associated with increased relapse and inferior overall survival. Multiple small molecule inhibitors of FLT3 signaling have been identified, two of which (midostaurin and gilteritinib) are currently approved in the United States, and many more of which are in clinical trials. Despite significant advances, resistance to FLT3 inhibitors through secondary FLT3 mutations, upregulation of parallel pathways, and extracellular signaling remains an ongoing challenge. Novel therapeutic strategies to overcome resistance, including combining FLT3 inhibitors with other antileukemic agents, development of new FLT3 inhibitors, and FLT3-directed immunotherapy are in active clinical development. Multiple questions regarding FLT3-mutated AML remain. In this review, we highlight several of the current most intriguing controversies in the field including the role of FLT3 inhibitors in maintenance therapy, the role of hematopoietic cell transplantation in FLT3-mutated AML, use of FLT3 inhibitors in FLT3 wild-type disease, significance of non-canonical FLT3 mutations, and finally, emerging concerns regarding clonal evolution.
Collapse
Affiliation(s)
- Vanessa E Kennedy
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Catherine C Smith
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
27
|
Roussel X, Daguindau E, Berceanu A, Desbrosses Y, Warda W, Neto da Rocha M, Trad R, Deconinck E, Deschamps M, Ferrand C. Acute Myeloid Leukemia: From Biology to Clinical Practices Through Development and Pre-Clinical Therapeutics. Front Oncol 2020; 10:599933. [PMID: 33363031 PMCID: PMC7757414 DOI: 10.3389/fonc.2020.599933] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
Recent studies have provided several insights into acute myeloid leukemia. Studies based on molecular biology have identified eight functional mutations involved in leukemogenesis, including driver and passenger mutations. Insight into Leukemia stem cells (LSCs) and assessment of cell surface markers have enabled characterization of LSCs from hematopoietic stem and progenitor cells. Clonal evolution has been described as having an effect similar to that of microenvironment alterations. Such biological findings have enabled the development of new targeted drugs, including drug inhibitors and monoclonal antibodies with blockage functions. Some recently approved targeted drugs have resulted in new therapeutic strategies that enhance standard intensive chemotherapy regimens as well as supportive care regimens. Besides the progress made in adoptive immunotherapy, since allogenic hematopoietic stem cell transplantation enabled the development of new T-cell transfer therapies, such as chimeric antigen receptor T-cell and transgenic TCR T-cell engineering, new promising strategies that are investigated.
Collapse
Affiliation(s)
- Xavier Roussel
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Etienne Daguindau
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Ana Berceanu
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Yohan Desbrosses
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Walid Warda
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | | | - Rim Trad
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | - Eric Deconinck
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Marina Deschamps
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| | - Christophe Ferrand
- Inserm EFS BFC, UMR1098 RIGHT, University Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
28
|
Jeon JY, Buelow DR, Garrison DA, Niu M, Eisenmann ED, Huang KM, Zavorka Thomas ME, Weber RH, Whatcott CJ, Warner SL, Orwick SJ, Carmichael B, Stahl E, Brinton LT, Lapalombella R, Blachly JS, Hertlein E, Byrd JC, Bhatnagar B, Baker SD. TP-0903 is active in models of drug-resistant acute myeloid leukemia. JCI Insight 2020; 5:140169. [PMID: 33268594 PMCID: PMC7714403 DOI: 10.1172/jci.insight.140169] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/16/2020] [Indexed: 12/17/2022] Open
Abstract
Effective treatment for AML is challenging due to the presence of clonal heterogeneity and the evolution of polyclonal drug resistance. Here, we report that TP-0903 has potent activity against protein kinases related to STAT, AKT, and ERK signaling, as well as cell cycle regulators in biochemical and cellular assays. In vitro and in vivo, TP-0903 was active in multiple models of drug-resistant FLT3 mutant AML, including those involving the F691L gatekeeper mutation and bone marrow microenvironment–mediated factors. Furthermore, TP-0903 demonstrated preclinical activity in AML models with FLT3-ITD and common co-occurring mutations in IDH2 and NRAS genes. We also showed that TP-0903 had ex vivo activity in primary AML cells with recurrent mutations including MLL-PTD, ASXL1, SRSF2, and WT1, which are associated with poor prognosis or promote clinical resistance to AML-directed therapies. Our preclinical studies demonstrate that TP-0903 is a multikinase inhibitor with potent activity against multiple drug-resistant models of AML that will have an immediate clinical impact in a heterogeneous disease like AML. TP-0903, a multikinase inhibitor, demonstrates preclinical activity in models of drug-resistant AML, including those involving FLT3 mutations, bone marrow microenvironment-mediated factors and recurrent mutations.
Collapse
Affiliation(s)
- Jae Yoon Jeon
- Division of Pharmaceutics and Pharmacology, College of Pharmacy
| | | | | | - Mingshan Niu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy
| | | | - Kevin M Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy
| | | | - Robert H Weber
- Division of Pharmaceutics and Pharmacology, College of Pharmacy
| | | | | | | | | | - Emily Stahl
- Division of Hematology, Department of Internal Medicine, and
| | | | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, and.,Comprehensive Cancer Center, The Ohio State University (OSU), Columbus, Ohio, USA
| | - James S Blachly
- Division of Hematology, Department of Internal Medicine, and.,Comprehensive Cancer Center, The Ohio State University (OSU), Columbus, Ohio, USA
| | - Erin Hertlein
- Division of Hematology, Department of Internal Medicine, and.,Comprehensive Cancer Center, The Ohio State University (OSU), Columbus, Ohio, USA
| | - John C Byrd
- Division of Pharmaceutics and Pharmacology, College of Pharmacy.,Division of Hematology, Department of Internal Medicine, and.,Comprehensive Cancer Center, The Ohio State University (OSU), Columbus, Ohio, USA
| | - Bhavana Bhatnagar
- Division of Hematology, Department of Internal Medicine, and.,Comprehensive Cancer Center, The Ohio State University (OSU), Columbus, Ohio, USA
| | - Sharyn D Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy.,Division of Hematology, Department of Internal Medicine, and.,Comprehensive Cancer Center, The Ohio State University (OSU), Columbus, Ohio, USA
| |
Collapse
|
29
|
Zabkiewicz J, Lazenby M, Edwards G, Bygrave CA, Omidvar N, Zhuang L, Knapper S, Guy C, Hills RK, Burnett AK, Alvares CL. Combination of a mitogen-activated protein kinase inhibitor with the tyrosine kinase inhibitor pacritinib combats cell adhesion-based residual disease and prevents re-expansion of FLT3-ITD acute myeloid leukaemia. Br J Haematol 2020; 191:231-242. [PMID: 32394450 DOI: 10.1111/bjh.16665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/23/2020] [Indexed: 01/18/2023]
Abstract
Minimal residual disease (MRD) in acute myeloid leukaemia (AML) poses a major challenge due to drug insensitivity and high risk of relapse. Intensification of chemotherapy and stem cell transplantation are often pivoted on MRD status. Relapse rates are high even with the integration of first-generation FMS-like tyrosine kinase 3 (FLT3) inhibitors in pre- and post-transplant regimes and as maintenance in FLT3-mutated AML. Pre-clinical progress is hampered by the lack of suitable modelling of residual disease and post-therapy relapse. In the present study, we investigated the nature of pro-survival signalling in primary residual tyrosine kinase inhibitor (TKI)-treated AML cells adherent to stroma and further determined their drug sensitivity in order to inform rational future therapy combinations. Using a primary human leukaemia-human stroma model to mimic the cell-cell interactions occurring in patients, the ability of several TKIs in clinical use, to abrogate stroma-driven leukaemic signalling was determined, and a synergistic combination with a mitogen-activated protein kinase (MEK) inhibitor identified for potential therapeutic application in the MRD setting. The findings reveal a common mechanism of stroma-mediated resistance that may be independent of mutational status but can be targeted through rational drug design, to eradicate MRD and reduce treatment-related toxicity.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Bridged-Ring Compounds/pharmacology
- Cell Adhesion/drug effects
- Child
- Child, Preschool
- Extracellular Signal-Regulated MAP Kinases
- Female
- Humans
- Infant
- Infant, Newborn
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Male
- Middle Aged
- Models, Biological
- Neoplasm, Residual
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/pharmacology
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- fms-Like Tyrosine Kinase 3/genetics
Collapse
Affiliation(s)
- Joanna Zabkiewicz
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| | - Michelle Lazenby
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| | - Gareth Edwards
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| | - Ceri A Bygrave
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| | - Nader Omidvar
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| | - Lihui Zhuang
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| | - Steve Knapper
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| | - Carol Guy
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| | - Robert K Hills
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| | - Alan K Burnett
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| | - Caroline L Alvares
- Academic Department of Haematology, University of Cardiff, H, eath Park, Cardiff, UK
| |
Collapse
|