1
|
Sanachai K, Hengphasatporn K, Chamni S, Suwanborirux K, Mahalapbutr P, Shigeta Y, Seetaha S, Choowongkomon K, Rungrotmongkol T. Exploring mimosamycin as a Janus kinase 2 inhibitor: A combined computational and experimental investigation. Comput Biol Chem 2025; 115:108346. [PMID: 39824143 DOI: 10.1016/j.compbiolchem.2025.108346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/20/2025]
Abstract
Janus kinases (JAKs) are a family of intracellular tyrosine kinases that play a crucial role in signal transduction pathways. JAK2 has been implicated in the pathogenesis of leukemia, making it a promising target for research aimed at reducing the risk of this disease. This study examined the potential of mimosamycin as a JAK2 inhibitor using both in vitro and in silico approaches. We performed a kinase assay to measure the IC50 of mimosamycin for JAK2 inhibition, which was found to be 22.52 ± 0.87 nM. Additionally, we utilized molecular docking, molecular dynamics simulations, and free energy calculations to investigate the inhibitory mechanism at the atomic level. Our findings revealed that mimosamycin interacts with JAK2 at several key regions: the hinge-conserved region (M929, Y931, L932, and G935), the G loop (L855 and V863), and the catalytic loop (L983). To enhance the binding affinity of mimosamycin toward JAK2, we designed derivatives with propanenitrile and cyclopentane substitutions on the naphthoquinone core structure. Notably, these newly designed analogs exhibited promising binding patterns against JAK2. These insights could aid in the rational development of novel JAK2 inhibitors, with potential applications in the treatment of leukemia and related diseases.
Collapse
Affiliation(s)
- Kamonpan Sanachai
- Center of Excellence in Structural and Computational Biology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kowit Hengphasatporn
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Supakarn Chamni
- Center of Excellent in Natural Products and Nanoparticles (NP2), Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Khanit Suwanborirux
- Center of Excellent in Natural Products and Nanoparticles (NP2), Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panupong Mahalapbutr
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Yasuteru Shigeta
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Supaphorn Seetaha
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand.
| | - Thanyada Rungrotmongkol
- Center of Excellence in Structural and Computational Biology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
2
|
Challoumas D, Simpson C, Arnold M, Mease P, Moots R, Ndosi M, Locher ZR. Janus-kinase inhibitor use in immune-mediated inflammatory diseases beyond licensed indications: A scoping review. Autoimmun Rev 2024; 24:103736. [PMID: 39743122 DOI: 10.1016/j.autrev.2024.103736] [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: 11/13/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
INTRODUCTION The use of Janus kinase inhibitors (JAKis) in immune-mediated inflammatory diseases (IMIDs) beyond licence is expanding rapidly. The aim of this scoping review was to identify and present the available evidence on the efficacy of JAKis in all conditions without marketing authorisation. METHODS Through a systematic literature search we identified studies including 5 or more patients that assessed the use of any JAKi for any efficacy outcome. Quantitative analyses in the form of pairwise meta-analyses were performed for eligible data from randomised controlled trials (RCTs) only. RESULTS Eighty-three (n = 83) studies in total were included in our review, assessing efficacy of JAKis in 34 IMIDs. In most conditions, JAKis exhibited generally positive effects, though the majority of evidence came from observational, non-comparative studies. Pairwise meta-analyses were possible for hidradenitis suppurativa and systemic lupus erythematosus (SLE). For hidradenitis suppurativa, we found a clear benefit of treatment with JAKis compared with placebo in achieving clinical response [OR 2.35, 95 % CI (1.24 to 4.46)]. For treatment-resistant SLE, the results were equivocal; JAKi showed some benefit over placebo but statistical significance was only reached for one of the two meta-analysed outcome measures [SLE Responder Index 4, OR 1.41, 95 % CI (1.01 to 1.98); SLE Disease Activity Index 2000; OR 1.36, 95 % CI (0.99 to 1.88)]. CONCLUSIONS There is a rapidly increasing use of JAKis beyond current licencing in most IMIDs. Large comparative trials are necessary to confirm efficacy and guide future licencing decisions.
Collapse
Affiliation(s)
- Dimitris Challoumas
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK; West of Scotland Trauma and Orthopaedic Training Programme, NHS Greater Glasgow and Clyde, Glasgow, UK.
| | - Cameron Simpson
- West of Scotland Trauma and Orthopaedic Training Programme, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Matthew Arnold
- West of Scotland Trauma and Orthopaedic Training Programme, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Philip Mease
- Rheumatology Research, Providence Swedish Medical Center and University of Washington, Seattle, USA
| | - Robert Moots
- Rheumatology Department, Aintree University Hospital, Liverpool University Hospitals NHS Trust and Faculty of Health, Care and Medicine, Edge Hill University, Ormskirk, UK
| | - Mwidimi Ndosi
- College of Health, Science and Society, University of the West of England, Bristol, UK
| | - Zoe Rutter Locher
- Rheumatology Department, Guy's and St Thomas' NHS Trust, London, United Kingdom
| |
Collapse
|
3
|
Namdaroglu S, Hidayet E, Aydin MS, Erkurt MA, Berber I, Cinar OE, Ozet G, Yilmaz S, Apaydin M, Dal MS, Korkmaz S, Basturk A, Ulaş T, Altuntas F. The role of ruxolitinib in the management of acute GVHD. Transfus Apher Sci 2024; 64:104055. [PMID: 39719750 DOI: 10.1016/j.transci.2024.104055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
BACKGROUND AND OBJECTIVES Following an allogeneic hematopoietic stem cell transplant (allo-HSCT), a primary cause of morbidity and mortality is still steroid-refractory acute graft-versus-host disease (SR-aGVHD). Recently, ruxolitinib, an oral inhibitor of JAK1 and JAK2, was approved for use in individuals suffering from SR-aGVHD. This study aimed to analyze the efficacy and toxicity of ruxolitinib in the real world. MATERIAL AND METHODS In the present study, we investigated the effectiveness and toxicity of ruxolitinib in patients with SR-aGVHD using a multicenter retrospective analysis. We enrolled 23 patients between 2018 and 2024 who received ruxolitinib treatment for SR-aGVHD. RESULTS The first response was acheived in a median of 28 days (range, 12-150). The overall response rate (ORR) for ruxolitinib therapy was 43.5 % (10/23) after one month and 61 % (14/23) after two months, respectively. The median overall survival was 69 months. Reactivation of cytomegalovirus (26.1 %) and grade 3-4 anemia (30.4 %) were the two main side effects of ruxolitinib therapy. Seven patients (30.4 %) passed away following a follow-up of a median of six months (range 1-70). The reasons for death included sepsis (n = 2, 28.6 %), progression of aGVHD (n = 3, 42.8 %), and other reasons. CONCLUSION Ruxolitinib has an ORR of 61 % for SR-aGVHD, making it a safe and effective therapy choice in real-world settings.
Collapse
Affiliation(s)
- Sinem Namdaroglu
- Dokuz Eylul University, Faculty of Medicine, Department of Hematology, Izmir, Turkey.
| | - Emine Hidayet
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey
| | - Muruvvet Seda Aydin
- Ankara Bilkent City Hospital, Department of Hematology and Bone Marrow Transplantation Unit, Ankara, Turkey
| | - Mehmet Ali Erkurt
- Inonu University, Faculty of Medicine, Department of Hematology, Malatya, Turkey
| | - Ilhami Berber
- Inonu University, Faculty of Medicine, Department of Hematology, Malatya, Turkey
| | - Olgu Erkin Cinar
- University of Health Sciences, Konya Medical Faculty, Department of Hematology and Bone Marrow Transplantation Unit, Konya, Turkey
| | - Gulsum Ozet
- Ankara Bilkent City Hospital, Department of Hematology and Bone Marrow Transplantation Unit, Ankara, Turkey
| | - Seda Yilmaz
- University of Health Sciences, Konya Medical Faculty, Department of Hematology and Bone Marrow Transplantation Unit, Konya, Turkey
| | - Merve Apaydin
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey
| | - Mehmet Sinan Dal
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey
| | - Serdal Korkmaz
- University of Health Sciences, Kayseri Medical Faculty, Department of Hematology and Bone Marrow Transplantation Unit, Kayseri, Turkey
| | - Abdulkadir Basturk
- University of Health Sciences, Konya Medical Faculty, Department of Hematology and Bone Marrow Transplantation Unit, Konya, Turkey
| | - Turgay Ulaş
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey
| | - Fevzi Altuntas
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey; Ankara Yildirim Beyazit University, School of Medicine, Department of Internal Medicine, Division of Hematology, Ankara, Turkey
| |
Collapse
|
4
|
Daoud S, Taha MO. Advances in the design and discovery of next-generation janus kinase-2 (JAK2) inhibitors for the treatment of myeloproliferative neoplasms. Expert Opin Drug Discov 2024; 19:1403-1415. [PMID: 39410824 DOI: 10.1080/17460441.2024.2417368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/13/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION Myeloproliferative neoplasms (MPNs) are rare hematopoietic disorders driven by mutations in the JAK-STAT signaling pathway genes. While JAK2 inhibitors have transformed MPN treatment, they do not eliminate the malignant clone or prevent disease progression in most patients. This limitation underscores the need for more effective therapies. AREA COVERED This review examines the evolution of JAK2 inhibitors for treating MPNs. Current JAK2 inhibitors primarily function as type I inhibitors, targeting the active kinase conformation, but their effectiveness is limited by ongoing JAK-STAT signaling. To overcome these limitations, next-generation therapies, such as type II JAK2 inhibitors and pseudokinase domain inhibitors, are being developed to target inactive kinase conformations and alternative signaling pathways. Furthermore, combination therapies with PI3K, mTOR, CDK4/6 inhibitors, and epigenetic modulators are being investigated for their potential synergistic effects, aiming for deeper and more durable responses in MPN patients. EXPERT OPINION Next-generation JAK2 inhibitors are needed to enhance current MPNs treatments by overcoming resistance, improving selectivity, targeting specific patient groups, and exploring combination therapies. Addressing challenges in drug design, preclinical testing, and clinical trials is crucial. Developing dual or multiple inhibitors targeting JAK2 and other MPN-related pathways is urgent to address complex signaling networks and improve efficacy.
Collapse
Affiliation(s)
- Safa Daoud
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Applied Sciences Private University, Amman, Jordan
| | - Mutasem Omar Taha
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, University of Jordan, Amman, Jordan
| |
Collapse
|
5
|
Pal R, Matada GSP, Teli G, Saha M, Patel R. Therapeutic potential of anticancer activity of nitrogen-containing heterocyclic scaffolds as Janus kinase (JAK) inhibitor: Biological activity, selectivity, and structure-activity relationship. Bioorg Chem 2024; 152:107696. [PMID: 39167870 DOI: 10.1016/j.bioorg.2024.107696] [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: 04/25/2024] [Revised: 07/21/2024] [Accepted: 08/04/2024] [Indexed: 08/23/2024]
Abstract
The JAK-STAT signalling pathway is primarily involved in cytokine signalling and induces various factors namely, erythropoietin, thrombopoietin, interferons, interleukins, and granulocyte colony-stimulating factors. These factors tremendously influenced understanding human health and illness, specifically cancer. Inhibiting the JAK/STAT pathway offers enormous therapeutic promises against cancer. Many JAK inhibitors are now being studied due to their efficacy in various cancer treatments. Further, the Nitrogen-heterocyclic (N-heterocyclic) scaffold has always shown to be a powerful tool for designing and discovering synthetic compounds with diverse pharmacological characteristics. The review focuses on several FDA-approved JAK inhibitors and their systematic categorization. The medicinal chemistry perspective is highlighted and classified review on the basis of N-heterocyclic molecules. Several examples of designing strategies of N-heterocyclic rings including pyrrolo-azepine, purine, 1H-pyrazolo[3,4-d]pyrimidine, 1H-pyrrolo[2,3-b]pyridine, pyrazole, thieno[3,2-d] pyrimidine, and, pyrimidine-based derivatives and their structure-activity relationships (SAR) are discussed. Among the various N-heterocyclic-based JAK inhibitors pyrimidine-containing compound 1 exhibited excellent inhibition activity against JAK2WT and mutated-JAK2V617F with IC50 of 2.01 and 18.84 nM respectively. Amino pyrimidine-containing compound 6 and thiopheno[3,2-d]pyrimidine-containing compound 13 expressed admirable JAK3 inhibition activity with IC50 of 1.7 nM and 1.38 nM respectively. Our review will support the medicinal chemists in refining and directing the development of novel N-heterocyclic-based JAK inhibitors.
Collapse
Affiliation(s)
- Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India.
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India.
| | - Ghanshyam Teli
- School of Pharmacy, Sangam University, Atoon, Bhilwara, 311001, Rajasthan, India
| | - Moumita Saha
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India; Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, MAHE, Karnataka
| | - Rajiv Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga-142001, Punjab, India
| |
Collapse
|
6
|
Ahmad N, Chen L, Yuan Z, Ma X, Yang X, Wang Y, Zhao Y, Jin H, Khaidamah N, Wang J, Lu J, Liu Z, Wu M, Wang Q, Qi Y, Wang C, Zhao Y, Piao Y, Huang R, Diao Y, Deng S, Shu X. Pyrimidine compounds BY4003 and BY4008 inhibit glioblastoma cells growth via modulating JAK3/STAT3 signaling pathway. Neurotherapeutics 2024; 21:e00431. [PMID: 39153914 PMCID: PMC11579875 DOI: 10.1016/j.neurot.2024.e00431] [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: 02/05/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024] Open
Abstract
Glioblastoma (GBM) is a brain tumor characterized by its aggressive and invasive properties. It is found that STAT3 is abnormally activated in GBM, and inhibiting STAT3 signaling can effectively suppress tumor progression. In this study, novel pyrimidine compounds, BY4003 and BY4008, were synthesized to target the JAK3/STAT3 signaling pathway, and their therapeutic efficacy and mechanisms of action were evaluated and compared with Tofacitinib in U251, A172, LN428 and patient-derived glioblastoma cells. The ADP-Glo™ kinase assay was utilized to assessed the inhibitory effects of BY4003 and BY4008 on JAK3, a crucial member of the JAK family. The results showed that both compounds significantly inhibited JAK3 enzyme activity, with IC50 values in the nanomolar range. The antiproliferative effects of BY4003, BY4008, and Tofacitinib on GBM and patient-derived glioblastoma cells were evaluated by MTT and H&E assays. The impact of BY4003 and BY4008 on GBM cell migration and apoptosis induction was assessed through wound healing, transwell, and TUNEL assays. STAT3-regulated protein expression and relative mRNA levels were analyzed by western blotting, immunocytochemistry, immunofluorescence, and qRT-PCR. It was found that BY4003, BY4008 and Tofacitinib could inhibit U251, A172, LN428 and patient-derived glioblastoma cells growth and proliferation. Results showed decreased expression of STAT3-associated proteins, including p-STAT3, CyclinD1, and Bcl-2, and increased expression of Bax, a pro-apoptotic protein, as well as significant down-regulation of STAT3 and STAT3-related genes. These findings suggested that BY4003 and BY4008 could inhibit GBM growth by suppressing the JAK3/STAT3 signaling pathway, providing valuable insights into the therapeutic development of GBM.
Collapse
Affiliation(s)
- Nisar Ahmad
- College of Pharmacy, Dalian Medical University, Dalian 116044, China; College of Basic Medical Science, Dalian Medical University, Dalian 116044, China; Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Lixue Chen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Zixi Yuan
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Xiaodong Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, China; Key Laboratories for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian 116044, China
| | - Xiaobo Yang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| | - Yinan Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China; Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, China; The First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Yongshun Zhao
- The First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Huan Jin
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Najib Khaidamah
- College of Pharmacy, Dalian Medical University, Dalian 116044, China; Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Jinan Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Jiashuo Lu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ziqi Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Moli Wu
- College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Qian Wang
- College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Chong Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yupu Zhao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yang Piao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Rujie Huang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China; Key Laboratories for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian 116044, China
| | - Sa Deng
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China; Institute of Integrative Medicine, Dalian Medical University, Dalian 116044, China; Key Laboratories for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
7
|
Shan M, Zhao X, Sun P, Qu X, Cheng G, Qin LP. Revisiting Structure-activity Relationships: Unleashing the potential of selective Janus kinase 1 inhibitors. Bioorg Chem 2024; 149:107506. [PMID: 38833989 DOI: 10.1016/j.bioorg.2024.107506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Janus kinases (JAKs), a kind of non-receptor tyrosine kinases, the function has been implicated in the regulation of cell proliferation, differentiation and apoptosis, immune, inflammatory response and malignancies. Among them, JAK1 represents an essential target for modulating cytokines involved in inflammation and immune function. Rheumatoid arthritis, atopic dermatitis, ulcerative colitis and psoriatic arthritis are areas where approved JAK1 drugs have been applied for the treatment. In the review, we provided a brief introduction to JAK1 inhibitors in market and clinical trials. The structures of high active JAK1 compounds (IC50 ≤ 0.1 nM) were highlighted, with primary focus on structure-activity relationship and selectivity. Moreover, the druggability processes of approved drugs and high active compounds were analyzed. In addition, the issues involved in JAK1 compounds clinical application as well as strategies to surmount these challenges, were discussed.
Collapse
Affiliation(s)
- Mengyi Shan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Xuan Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Peng Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Xinhao Qu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Gang Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China.
| | - Lu-Ping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China.
| |
Collapse
|
8
|
Tefferi A, Pardanani A. Momelotinib for myelofibrosis: our 14 years of experience with 100 clinical trial patients and recent FDA approval. Blood Cancer J 2024; 14:47. [PMID: 38499521 PMCID: PMC10948874 DOI: 10.1038/s41408-024-01029-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/20/2024] [Accepted: 03/05/2024] [Indexed: 03/20/2024] Open
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Animesh Pardanani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
9
|
Pessino G, Scotti C, Maggi M, Immuno-Hub Consortium. Hepatocellular Carcinoma: Old and Emerging Therapeutic Targets. Cancers (Basel) 2024; 16:901. [PMID: 38473265 DOI: 10.3390/cancers16050901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Liver cancer, predominantly hepatocellular carcinoma (HCC), globally ranks sixth in incidence and third in cancer-related deaths. HCC risk factors include non-viral hepatitis, alcohol abuse, environmental exposures, and genetic factors. No specific genetic alterations are unequivocally linked to HCC tumorigenesis. Current standard therapies include surgical options, systemic chemotherapy, and kinase inhibitors, like sorafenib and regorafenib. Immunotherapy, targeting immune checkpoints, represents a promising avenue. FDA-approved checkpoint inhibitors, such as atezolizumab and pembrolizumab, show efficacy, and combination therapies enhance clinical responses. Despite this, the treatment of hepatocellular carcinoma (HCC) remains a challenge, as the complex tumor ecosystem and the immunosuppressive microenvironment associated with it hamper the efficacy of the available therapeutic approaches. This review explores current and advanced approaches to treat HCC, considering both known and new potential targets, especially derived from proteomic analysis, which is today considered as the most promising approach. Exploring novel strategies, this review discusses antibody drug conjugates (ADCs), chimeric antigen receptor T-cell therapy (CAR-T), and engineered antibodies. It then reports a systematic analysis of the main ligand/receptor pairs and molecular pathways reported to be overexpressed in tumor cells, highlighting their potential and limitations. Finally, it discusses TGFβ, one of the most promising targets of the HCC microenvironment.
Collapse
Affiliation(s)
- Greta Pessino
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Claudia Scotti
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Maristella Maggi
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Immuno-Hub Consortium
- Unit of Immunology and General Pathology, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
10
|
Tefferi A, Pardanani A, Gangat N. Momelotinib expands the therapeutic armamentarium for myelofibrosis: Impact on hierarchy of treatment choices. Am J Hematol 2024; 99:300-308. [PMID: 38164985 DOI: 10.1002/ajh.27163] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/05/2023] [Indexed: 01/03/2024]
Abstract
The primary objective of treatment in myelofibrosis (MF) is prolongation of life, which is currently accomplished only by allogeneic hematopoietic stem cell transplantation (AHSCT). Determination of optimal timing for AHSCT is facilitated by molecular risk stratification. Non-transplant treatment options in MF are palliative in scope and include Janus kinase 2 (JAK2) inhibitors (JAKi): momelotinib (FDA approved on September 15, 2023), ruxolitinib (November 16, 2011), fedratinib (August 16, 2019), and pacritinib (February 28, 2022); all four JAKi are effective in reducing spleen size and alleviating symptoms, considered a drug class effect and attributed to their canonical JAK-STAT inhibitory mechanism of action. In addition, momelotinib exhibits erythropoietic effect, attributed to alleviation of ineffective erythropoiesis through inhibition of activin A receptor type-I (ACVR1). In transplant-ineligible or deferred patients, the order of treatment preference is based on specific symptoms and individual assessment of risk tolerance. Because of drug-induced immunosuppression and other toxicities attributed to JAKi, we prefer non-JAKi drugs as initial treatment for MF-associated anemia that is not accompanied by treatment-requiring splenomegaly or constitutional symptoms. Otherwise, it is reasonable to consider momelotinib as the first-line JAKi treatment of choice, in order to target the triad of quality-of-life offenders in MF: anemia, splenomegaly, and constitutional symptoms/cachexia. For second-line therapy, we favor ruxolitinib, over fedratinib, based on toxicity profile. Pacritinib and fedratinib provide alternative options in the presence of severe thrombocytopenia or ruxolitinib-resistance/intolerance, respectively. Splenectomy remains a viable option for drug-resistant symptomatic splenomegaly and cytopenia.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Animesh Pardanani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Naseema Gangat
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
11
|
Liang D, Wang Q, Zhang W, Tang H, Song C, Yan Z, Liang Y, Wang H. JAK/STAT in leukemia: a clinical update. Mol Cancer 2024; 23:25. [PMID: 38273387 PMCID: PMC10811937 DOI: 10.1186/s12943-023-01929-1] [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: 11/01/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
Over the past three decades, considerable efforts have been expended on understanding the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway in leukemia, following the identification of the JAK2V617F mutation in myeloproliferative neoplasms (MPNs). The aim of this review is to summarize the latest progress in our understanding of the involvement of the JAK/STAT signaling pathway in the development of leukemia. We also attempt to provide insights into the current use of JAK/STAT inhibitors in leukemia therapy and explore pertinent clinical trials in this field.
Collapse
Affiliation(s)
- Dong Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qiaoli Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wenbiao Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Cailu Song
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhimin Yan
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
| | - Yang Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Hua Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
12
|
Park E, Park S, Lee SJ, Jeong D, Jin H, Moon H, Cha B, Kim D, Ma S, Seo W, Han SH, Lee YS, Kang S. Identification and Biological Evaluation of a Potent and Selective JAK1 Inhibitor for the Treatment of Pulmonary Fibrosis. J Med Chem 2023; 66:16342-16363. [PMID: 38031930 DOI: 10.1021/acs.jmedchem.3c01712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Janus kinase 1 (JAK1) plays a pivotal role in regulating inflammation and fibrosis via the JAK/STAT signaling pathway, making it a promising target for associated diseases. In this study, we explored the modification of an N-methyl 1H-pyrrolo[2,3-b]pyridine-5-carboxylate core, leading to the identification of 4-(((2S,4S)-1-(4-trifluoromethyl)-2-methylpiperidin-4-yl)amino)-N-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (36b) as a highly potent and selective JAK1 inhibitor. Compound 36b exhibited an impressive IC50 value of 0.044 nM for JAK1 and demonstrated remarkable selectivity of 382-fold, 210-fold, and 1325-fold specificity over JAK2, JAK3, and TYK2, respectively. The kinase panel assays further confirmed its specificity, and cell-based experiments established its efficacy in inhibiting JAK1-STAT phosphorylation in human L-132 or SK-MES-1 cells. Pharmacokinetic studies revealed that compound 36b boasts an oral bioavailability exceeding 36%. In a bleomycin-induced fibrosis mouse model, compound 36b significantly reduced STAT3 phosphorylation, resulting in improvement in body weight and reduced collagen deposition, all achieved without significant side effects.
Collapse
Affiliation(s)
- Eunsun Park
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seolhee Park
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sun Joo Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea
| | - Dayeon Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hee Jin
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Heegyum Moon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea
| | - Boksik Cha
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea
| | - Dayea Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea
| | - Seonghee Ma
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Wonhyo Seo
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seung-Hee Han
- Central Research Laboratory, KOREA PHARMA Co. Ltd., Jeyakgongdan 3-gil, Hyangnam-eup, Hwaseong-si, Gyeonggi-do 16630, Republic of Korea
| | - Yun-Sil Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Soosung Kang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
13
|
Chung C. Current therapies for classic myeloproliferative neoplasms: A focus on pathophysiology and supportive care. Am J Health Syst Pharm 2023; 80:1624-1636. [PMID: 37556726 DOI: 10.1093/ajhp/zxad181] [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: 08/08/2023] [Indexed: 08/11/2023] Open
Abstract
PURPOSE This article concisely evaluates current therapies that have received regulatory approval for the treatment of classic myeloproliferative neoplasms (MPNs). Pertinent pathophysiology and supportive care are discussed. Emerging therapies are also briefly described. SUMMARY MPNs are a heterogeneous group of diseases characterized by acquired abnormalities of hematopoietic stem cells (HSCs), resulting in the generation of transformed myeloid progenitor cells that overproduce mature and immature cells within the myeloid lineage. Mutations in JAK2 and other driver oncogenes are central to the genetic variability of these diseases. Cytoreductive therapies such as hydroxyurea, anagrelide, interferon, and therapeutic phlebotomy aim to lower the risk of thrombotic events without exposing patients to an increased risk of leukemic transformation. However, no comparisons can be made between these therapies, as reduction of thrombotic risk has not been used as an endpoint. On the other hand, Janus kinase (JAK) inhibitors such as ruxolitinib, fedratinib, pacritinib, and momelotinib (an investigational agent at the time of writing) directly target the constitutively activated JAK-signal transducer and activator of transcription (JAK-STAT) pathway of HSCs in the bone marrow. Mutations of genes in the JAK-STAT signaling pathway provide a unifying understanding of MPNs, spur therapeutic innovations, and represent opportunities for pharmacists to optimize mitigation strategies for both disease-related and treatment-related adverse effects. CONCLUSION Treatment options for MPNs span a wide range of disease mechanisms. The growth of targeted therapies holds promise for expanding the treatment arsenal for these rare, yet complex diseases and creates opportunities to optimize supportive care for affected patients.
Collapse
|
14
|
Tefferi A. Jaktinib and momelotinib for the treatment of myelofibrosis-Birds of a feather? Am J Hematol 2023; 98:1517-1519. [PMID: 37471656 DOI: 10.1002/ajh.27036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
15
|
Seetharam SM, Liu Y, Wu J, Fechter L, Murugesan K, Maecker H, Gotlib J, Zehnder J, Paulmurugan R, Krishnan A. Enkurin: a novel marker for myeloproliferative neoplasms from platelet, megakaryocyte, and whole blood specimens. Blood Adv 2023; 7:5433-5445. [PMID: 37315179 PMCID: PMC10509670 DOI: 10.1182/bloodadvances.2022008939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/10/2023] [Accepted: 05/30/2023] [Indexed: 06/16/2023] Open
Abstract
Impaired protein homeostasis, though well established in age-related disorders, has been recently linked with the pathogenesis of myeloproliferative neoplasms (MPNs). However, little is known about MPN-specific modulators of proteostasis, thus impeding our ability for increased mechanistic understanding and discovery of additional therapeutic targets. Loss of proteostasis, in itself, is traced to dysregulated mechanisms in protein folding and intracellular calcium signaling at the endoplasmic reticulum (ER). Here, using ex vivo and in vitro systems (including CD34+ cultures from patient bone marrow and healthy cord/peripheral blood specimens), we extend our prior data from platelet RNA sequencing in patients with MPN and discover select proteostasis-associated markers at RNA and/or protein levels in each of platelet, parent megakaryocyte, and whole blood specimens. Importantly, we identify a novel role in MPNs for enkurin (ENKUR), a calcium mediator protein originally implicated only in spermatogenesis. Our data reveal consistent ENKUR downregulation at both RNA and protein levels across specimens from patients with MPN and experimental models (including upon treatment with thapsigargin, an agent that causes protein misfolding in the ER by selective loss of calcium), with a concomitant upregulation of a cell cycle marker, CDC20. Silencing of ENKUR using short hairpin RNA in CD34+-derived megakaryocytes further confirms this association with CDC20 at both RNA and protein levels and indicates a likely role for the PI3K/Akt pathway. Together, our work sheds light on enkurin as a novel marker of MPN pathogenesis and indicates further mechanistic investigation into a role for dysregulated calcium homeostasis and ER and protein folding stress in MPN transformation.
Collapse
Affiliation(s)
| | - Yi Liu
- Department of Radiology, Stanford University, Stanford, CA
| | - Jason Wu
- High-Throughput Bioscience Center and Stanford Genomics, Stanford University School of Medicine, Stanford, CA
| | - Lenn Fechter
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | | | - Holden Maecker
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA
| | - Jason Gotlib
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - James Zehnder
- Department of Pathology, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Anandi Krishnan
- Department of Pathology, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
16
|
Green JR, Mahalingaiah PKS, Gopalakrishnan SM, Liguori MJ, Mittelstadt SW, Blomme EAG, Van Vleet TR. Off-target pharmacological activity at various kinases: Potential functional and pathological side effects. J Pharmacol Toxicol Methods 2023; 123:107468. [PMID: 37553032 DOI: 10.1016/j.vascn.2023.107468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/16/2023] [Accepted: 08/01/2023] [Indexed: 08/10/2023]
Abstract
In drug discovery, during the lead optimization and candidate characterization stages, novel small molecules are frequently evaluated in a battery of in vitro pharmacology assays to identify potential unintended, off-target interactions with various receptors, transporters, ion channels, and enzymes, including kinases. Furthermore, these screening panels may also provide utility at later stages of development to provide a mechanistic understanding of unexpected safety findings. Here, we present a compendium of the most likely functional and pathological outcomes associated with interaction(s) to a panel of 95 kinases based on an extensive curation of the scientific literature. This panel of kinases was designed by AbbVie based on safety-related data extracted from the literature, as well as from over 20 years of institutional knowledge generated from discovery efforts. For each kinase, the scientific literature was reviewed using online databases and the most often reported functional and pathological effects were summarized. This work should serve as a practical guide for small molecule drug discovery scientists and clinical investigators to predict and/or interpret adverse effects related to pharmacological interactions with these kinases.
Collapse
Affiliation(s)
- Jonathon R Green
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States.
| | | | - Sujatha M Gopalakrishnan
- Drug Discovery Science and Technology, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Michael J Liguori
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Scott W Mittelstadt
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Eric A G Blomme
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Terry R Van Vleet
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| |
Collapse
|
17
|
Nair PC, Piehler J, Tvorogov D, Ross DM, Lopez AF, Gotlib J, Thomas D. Next-Generation JAK2 Inhibitors for the Treatment of Myeloproliferative Neoplasms: Lessons from Structure-Based Drug Discovery Approaches. Blood Cancer Discov 2023; 4:352-364. [PMID: 37498362 PMCID: PMC10472187 DOI: 10.1158/2643-3230.bcd-22-0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 04/20/2023] [Accepted: 06/07/2023] [Indexed: 07/28/2023] Open
Abstract
Selective inhibitors of Janus kinase (JAK) 2 have been in demand since the discovery of the JAK2 V617F mutation present in patients with myeloproliferative neoplasms (MPN); however, the structural basis of V617F oncogenicity has only recently been elucidated. New structural studies reveal a role for other JAK2 domains, beyond the kinase domain, that contribute to pathogenic signaling. Here we evaluate the structure-based approaches that led to recently-approved type I JAK2 inhibitors (fedratinib and pacritinib), as well as type II (BBT594 and CHZ868) and pseudokinase inhibitors under development (JNJ7706621). With full-length JAK homodimeric structures now available, superior selective and mutation-specific JAK2 inhibitors are foreseeable. SIGNIFICANCE The JAK inhibitors currently used for the treatment of MPNs are effective for symptom management but not for disease eradication, primarily because they are not strongly selective for the mutant clone. The rise of computational and structure-based drug discovery approaches together with the knowledge of full-length JAK dimer complexes provides a unique opportunity to develop better targeted therapies for a range of conditions driven by pathologic JAK2 signaling.
Collapse
Affiliation(s)
- Pramod C. Nair
- Cancer Program, South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide, Adelaide, Australia
- Discipline of Medicine, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- Discipline of Clinical Pharmacology, Flinders Health and Medical Research Institute (FHMRI) Cancer Program, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Jacob Piehler
- Department of Biology and Center of Cellular Nanoanalytics, University of Osnabrück, Osnabrück, Germany
| | - Denis Tvorogov
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - David M. Ross
- Cancer Program, South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide, Adelaide, Australia
- Discipline of Medicine, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
- Department of Hematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Angel F. Lopez
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| | - Jason Gotlib
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Daniel Thomas
- Cancer Program, South Australian Health and Medical Research Institute (SAHMRI), University of Adelaide, Adelaide, Australia
- Discipline of Medicine, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- Department of Hematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
18
|
Duminuco A, Vetro C, Giallongo C, Palumbo GA. The pharmacotherapeutic management of patients with myelofibrosis: looking beyond JAK inhibitors. Expert Opin Pharmacother 2023; 24:1449-1461. [PMID: 37341682 DOI: 10.1080/14656566.2023.2228695] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/20/2023] [Indexed: 06/22/2023]
Abstract
INTRODUCTION The approach to myelofibrosis (MF) has been revolutionized in recent years, overcoming the traditional therapies, often not very effective. Janus kinase inhibitors (JAKi - from ruxolitinib up to momelotinib) were the first class of drugs with considerable results. AREAS COVERED Ongoing, new molecules are being tested that promise to give hope even to those patients not eligible for bone marrow transplants who become intolerant or are refractory to JAKi, for which therapeutic hopes are currently limited. Telomerase, murine double minute 2 (MDM2), phosphatidylinositol 3-kinase δ (PI3Kδ), BCL-2/xL, and bromodomain and extra-terminal motif (BET) inhibitors are the drugs with promising results in clinical trials and close to closure with consequent placing on the market, finally allowing JAK to look beyond. The novelty of the MF field was searched in the PubMed database, and the recently completed/ongoing trials are extrapolated from the ClinicalTrial website. EXPERT OPINION From this point of view, the use of new molecules widely described in this review, probably in association with JAKi, will represent the future treatment of choice in MF, leaving, in any case, the potential new approaches actually in an early stage of development, such as the use of immunotherapy in targeting CALR, which is coming soon.
Collapse
Affiliation(s)
- Andrea Duminuco
- Hematology with BMT Unit, A.O.U. "G. Rodolico-San Marco", Catania, Italy
| | - Calogero Vetro
- Hematology with BMT Unit, A.O.U. "G. Rodolico-San Marco", Catania, Italy
| | - Cesarina Giallongo
- Dipartimento di Scienze Mediche Chirurgiche E Tecnologie Avanzate "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Giuseppe Alberto Palumbo
- Hematology with BMT Unit, A.O.U. "G. Rodolico-San Marco", Catania, Italy
- Dipartimento di Scienze Mediche Chirurgiche E Tecnologie Avanzate "G.F. Ingrassia", University of Catania, Catania, Italy
| |
Collapse
|
19
|
Verstovsek S, Mesa RA, Livingston RA, Hu W, Mascarenhas J. Ten years of treatment with ruxolitinib for myelofibrosis: a review of safety. J Hematol Oncol 2023; 16:82. [PMID: 37501130 PMCID: PMC10373260 DOI: 10.1186/s13045-023-01471-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/22/2023] [Indexed: 07/29/2023] Open
Abstract
Myelofibrosis (MF) is a chronic myeloproliferative neoplasm characterized by bone marrow fibrosis, anemia, extramedullary hematopoiesis, and splenomegaly. Patients with MF are at risk for reduced survival versus the general population and often experience burdensome signs and symptoms that reduce quality of life. The oral Janus kinase (JAK) 1/JAK2 inhibitor ruxolitinib was initially approved by the US Food and Drug Administration in 2011 for the treatment of patients with intermediate or high-risk MF, including primary MF, post-polycythemia vera MF, and post-essential thrombocythemia MF, based on efficacy and safety findings from the randomized, controlled, phase 3 COMFORT trials. Over a decade later, ruxolitinib continues to be the standard of care in higher-risk MF, and dose optimization and management remain crucial for safely maximizing clinical benefits of ruxolitinib. This review summarizes the safety profile of ruxolitinib in patients with MF in the COMFORT trials leading up to approval and in the subsequent JUMP, ROBUST, EXPAND, and REALISE trials; in pooled analyses; and in postmarketing analyses in the 10 years following approval. There is a focus on the occurrence of common hematologic and nonhematologic adverse events, with guidance provided on the management of patients with anemia or thrombocytopenia, including dosing strategies based on findings from the REALISE and EXPAND trials. Finally, to ensure a greater understanding of the safety profile of ruxolitinib, practical considerations are discussed.
Collapse
Affiliation(s)
- Srdan Verstovsek
- Leukemia Department, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Ruben A Mesa
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Medical Center Blvd, 11th Floor, Winston-Salem, NC, 27157, USA
| | | | - Wilson Hu
- Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, DE, 19803, USA
| | - John Mascarenhas
- Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| |
Collapse
|
20
|
Kim BK, Eah KY, Park JM. Essential Thrombocythemia and Ischemic Stroke: A Case Series of Five JAK2-Positive Patients. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1300. [PMID: 37512112 PMCID: PMC10384755 DOI: 10.3390/medicina59071300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023]
Abstract
Background and Objectives: Essential thrombocythemia (ET) is a chronic myeloproliferative neoplasm characterized by elevated platelet counts and an increased risk of thrombotic events, including ischemic strokes. Materials and Methods: We conducted a retrospective analysis of data from consecutive ischemic stroke patients with ET between March 2014 and February 2023. Results: This case series describes the clinical presentation, radiological features, and management of five patients with ET-associated ischemic strokes, all harboring the JAK2 mutation. The diverse radiological findings suggest that both large and small vessel diseases may be influenced by the prothrombotic state induced by ET. A significant elevation in platelet count was observed to correlate with the emergence of new acute infarctions in some cases. Conclusions: The study highlights combined use of antiplatelet and cytoreductive therapy in preventing secondary stroke events in patients with ET and JAK2 mutations. The heterogeneity of stroke patterns in this population necessitates a comprehensive understanding of the underlying pathophysiological mechanisms and tailored therapeutic approaches.
Collapse
Affiliation(s)
- Byong-Kyu Kim
- Division of Cardiology, Department of Internal Medicine, Dongguk University Gyeongju Hospital, Dongguk University College of Medicine, Gyeongju 38067, Republic of Korea
| | - Kyung Yoon Eah
- Department of Neurology, Dongguk University Gyeongju Hospital, Dongguk University College of Medicine, Gyeongju 38067, Republic of Korea
| | - Jin-Mo Park
- Department of Neurology, Dongguk University Gyeongju Hospital, Dongguk University College of Medicine, Gyeongju 38067, Republic of Korea
| |
Collapse
|
21
|
Courdy C, Platteeuw L, Ducau C, De Araujo I, Boet E, Sahal A, Saland E, Edmond V, Tavitian S, Bertoli S, Cougoul P, Granat F, Poillet L, Marty C, Plo I, Sarry JE, Manenti S, Mansat-De Mas V, Joffre C. Targeting PP2A-dependent autophagy enhances sensitivity to ruxolitinib in JAK2 V617F myeloproliferative neoplasms. Blood Cancer J 2023; 13:106. [PMID: 37423955 DOI: 10.1038/s41408-023-00875-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Abstract
The Janus kinase 2 (JAK2)-driven myeloproliferative neoplasms (MPNs) are chronic malignancies associated with high-risk complications and suboptimal responses to JAK inhibitors such as ruxolitinib. A better understanding of cellular changes induced by ruxolitinib is required to develop new combinatory therapies to improve treatment efficacy. Here, we demonstrate that ruxolitinib induced autophagy in JAK2V617F cell lines and primary MPN patient cells through the activation of protein phosphatase 2A (PP2A). Inhibition of autophagy or PP2A activity along with ruxolitinib treatment reduced proliferation and increased the death of JAK2V617F cells. Accordingly, proliferation and clonogenic potential of JAK2V617F-driven primary MPN patient cells, but not of normal hematopoietic cells, were markedly impaired by ruxolitinib treatment with autophagy or PP2A inhibitor. Finally, preventing ruxolitinib-induced autophagy with a novel potent autophagy inhibitor Lys05 improved leukemia burden reduction and significantly prolonged the mice's overall survival compared with ruxolitinib alone. This study demonstrates that PP2A-dependent autophagy mediated by JAK2 activity inhibition contributes to resistance to ruxolitinib. Altogether, our data support that targeting autophagy or its identified regulator PP2A could enhance sensitivity to ruxolitinib of JAK2V617F MPN cells and improve MPN patient care.
Collapse
Affiliation(s)
- Charly Courdy
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
| | - Loïc Platteeuw
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
| | - Charlotte Ducau
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
| | - Isabelle De Araujo
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
| | - Emeline Boet
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
| | - Ambrine Sahal
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
| | - Estelle Saland
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
| | - Valérie Edmond
- INSERM UMR1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Suzanne Tavitian
- Service d'hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Université de Toulouse III Paul Sabatier, Toulouse, France
| | - Sarah Bertoli
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
- Service d'hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Université de Toulouse III Paul Sabatier, Toulouse, France
| | - Pierre Cougoul
- Service de médecine interne, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Fanny Granat
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
| | - Laura Poillet
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
| | - Caroline Marty
- INSERM UMR1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Isabelle Plo
- INSERM UMR1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France
| | - Stéphane Manenti
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France
| | - Véronique Mansat-De Mas
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France.
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France.
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole, Université de Toulouse III Paul Sabatier, Toulouse, France.
| | - Carine Joffre
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037, CNRS UMR 5071, Université de Toulouse, Toulouse, France.
- Equipe labellisée La Ligue contre le Cancer 2018, Toulouse, France.
| |
Collapse
|
22
|
Faida P, Attiogbe MKI, Majeed U, Zhao J, Qu L, Fan D. Lung cancer treatment potential and limits associated with the STAT family of transcription factors. Cell Signal 2023:110797. [PMID: 37423343 DOI: 10.1016/j.cellsig.2023.110797] [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: 04/06/2023] [Revised: 06/19/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
Lung cancer is one of the mortal cancers and the leading cause of cancer-related mortality, with a cancer survival rate of fewer than 5% in developing nations. This low survival rate can be linked to things like late-stage detection, quick postoperative recurrences in patients receiving therapy, and chemoresistance developing against various lung cancer treatments. Signal transducer and activator of transcription (STAT) family of transcription factors are involved in lung cancer cell proliferation, metastasis, immunological control, and treatment resistance. By interacting with specific DNA sequences, STAT proteins trigger the production of particular genes, which in turn result in adaptive and incredibly specific biological responses. In the human genome, seven STAT proteins have been discovered (STAT1 to STAT6, including STAT5a and STAT5b). Many external signaling proteins can activate unphosphorylated STATs (uSTATs), which are found inactively in the cytoplasm. When STAT proteins are activated, they can increase the transcription of several target genes, which leads to unchecked cellular proliferation, anti-apoptotic reactions, and angiogenesis. The effects of STAT transcription factors on lung cancer are variable; some are either pro- or anti-tumorigenic, while others maintain dual, context-dependent activities. Here, we give a succinct summary of the various functions that each member of the STAT family plays in lung cancer and go into more detail about the advantages and disadvantages of pharmacologically targeting STAT proteins and their upstream activators in the context of lung cancer treatment.
Collapse
Affiliation(s)
- Paison Faida
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Mawusse K I Attiogbe
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Usman Majeed
- College of Food Science and Technology, Northwest University, Xi'an, Shaanxi 710069, China
| | - Jing Zhao
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Linlin Qu
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech. & Biomed. Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
23
|
Arwood ML, Liu Y, Harkins SK, Weinstock DM, Yang L, Stevenson KE, Plana OD, Dong J, Cirka H, Jones KL, Virtanen AT, Gupta DG, Ceas A, Lawney B, Yoda A, Leahy C, Hao M, He Z, Choi HG, Wang Y, Silvennoinen O, Hubbard SR, Zhang T, Gray NS, Li LS. New scaffolds for type II JAK2 inhibitors overcome the acquired G993A resistance mutation. Cell Chem Biol 2023; 30:618-631.e12. [PMID: 37290440 PMCID: PMC10495080 DOI: 10.1016/j.chembiol.2023.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 02/07/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023]
Abstract
Recurrent JAK2 alterations are observed in myeloproliferative neoplasms, B-cell acute lymphoblastic leukemia, and other hematologic malignancies. Currently available type I JAK2 inhibitors have limited activity in these diseases. Preclinical data support the improved efficacy of type II JAK2 inhibitors, which lock the kinase in the inactive conformation. By screening small molecule libraries, we identified a lead compound with JAK2 selectivity. We highlight analogs with on-target biochemical and cellular activity and demonstrate in vivo activity using a mouse model of polycythemia vera. We present a co-crystal structure that confirms the type II binding mode of our compounds with the "DFG-out" conformation of the JAK2 activation loop. Finally, we identify a JAK2 G993A mutation that confers resistance to the type II JAK2 inhibitor CHZ868 but not to our analogs. These data provide a template for identifying novel type II kinase inhibitors and inform further development of agents targeting JAK2 that overcome resistance.
Collapse
Affiliation(s)
- Matthew L Arwood
- Molecular and Translational Cancer Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Yao Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Shannon K Harkins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - David M Weinstock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Cancer Biology Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Biological and Biomedical Sciences Program, Harvard Medical School, Boston, MA 02115, USA
| | - Lei Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kristen E Stevenson
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Olivia D Plana
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jingyun Dong
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA; Skirball Institute of Biomolecular Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Haley Cirka
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kristen L Jones
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Anniina T Virtanen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Dikshat G Gupta
- Molecular and Translational Cancer Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Amanda Ceas
- Molecular and Translational Cancer Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Brian Lawney
- Center for Cancer Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Akinori Yoda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Catharine Leahy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Mingfeng Hao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Zhixiang He
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Hwan Geun Choi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Yaning Wang
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Olli Silvennoinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Stevan R Hubbard
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA; Skirball Institute of Biomolecular Medicine, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Tinghu Zhang
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Loretta S Li
- Molecular and Translational Cancer Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Division of Hematology, Oncology, and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
24
|
Alsuhibani AS, Alharthi RM, AlSuhaymi S, Alnahdi MA, Almohideb M. Prescription Pattern of Tofacitinib for Alopecia Areata Among the Dermatologists in Saudi Arabia: A Cross-Sectional Study. Cureus 2023; 15:e40445. [PMID: 37325685 PMCID: PMC10267882 DOI: 10.7759/cureus.40445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction Alopecia areata (AA) is a complex autoimmune condition that causes nonscarring hair loss. In Saudi Arabia, AA accounts for 1-2% of new dermatological outpatient visits. It typically presents with sharply demarcated round patches of hair loss and may present at any age. Traditional medical therapies include corticosteroids and immunotherapy. Choosing the ideal treatment depends on multiple factors such as patient age, disease severity, efficacy, side effects, and remission rate. Recent medications that have been used for treating AA are Janus kinase inhibitors. Aim The aim of the study is to assess the awareness and attitude of dermatologists and their use of Tofacitinib in treating AA. Method A cross-sectional study was conducted in 2019 across 14 major cities in Saudi Arabia. A self-administered online questionnaire was specifically developed and used. Dermatologists from government hospitals and private clinics were included through non-probability convenience sampling. The collected data was entered into Microsoft Excel and analyzed using SPSS program version 24. Results In total, out of 546 Dermatologists across Saudi Arabia who responded to the questionnaire, 127 (23.2%) physicians prescribed Tofacitinib in their practice. Out of those who prescribed the drug for AA cases, 58 dermatologists (45.6%) prescribed Tofacitinib after the failure of steroid injections. Among the 127 dermatologists who have utilized Tofacitinib in their practice, 92 (72.4%) believe that Tofacitinib is effective in treating AA. Almost 200 (47.7%) Dermatologists who never prescribed Tofacitinib reported that the main reason was due to the unavailability of the drug in the clinic they were practicing. Conclusions To conclude, out of 546 dermatologists working in Saudi Arabia, 127 (23.2%) prescribe Tofacitinib to treat AA. Ninety-two (72.4%) of the participants reported the effectiveness of Tofacitinib. Two hundred (47.7%) dermatologists who never prescribe Tofacitinib reported that the main reason was due to the unavailability. However, this would raise the need for more research regarding JAK inhibitors generally and Tofacitinib specifically, focusing on the effectiveness versus the side effects of Tofacitinib.
Collapse
Affiliation(s)
- Abdulaziz S Alsuhibani
- Collage of Medicine, King Saud Bin Abdulaziz University for Health Sciences College of Medicine, Riyadh, SAU
| | | | - Saba AlSuhaymi
- Department of Dermatology, King Faisal Specialist Hospital and Research Centre, Riyadh, SAU
| | - Muhannad A Alnahdi
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, CAN
| | | |
Collapse
|
25
|
Beckman JD, DaSilva A, Aronovich E, Nguyen A, Nguyen J, Hargis G, Reynolds D, Vercellotti GM, Betts B, Wood DK. JAK-STAT inhibition reduces endothelial prothrombotic activation and leukocyte-endothelial proadhesive interactions. J Thromb Haemost 2023; 21:1366-1380. [PMID: 36738826 PMCID: PMC10246778 DOI: 10.1016/j.jtha.2023.01.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Vascular activation is characterized by increased proinflammatory, pro thrombotic, and proadhesive signaling. Several chronic and acute conditions, including Bcr-abl-negative myeloproliferative neoplasms (MPNs), graft-vs-host disease, and COVID-19 have been noted to have increased activation of the janus kinase (JAK)-signal transducer and downstream activator of transcription (STAT) pathways. Two notable inhibitors of the JAK-STAT pathway are ruxolitinib (JAK1/2 inhibitor) and fedratinib (JAK2 inhibitor), which are currently used to treat MPN patients. However, in some conditions, it has been noted that JAK inhibitors can increase the risk of thromboembolic complications. OBJECTIVES We sought to define the anti-inflammatory and antithrombotic effects of JAK-STAT inhibitors in vascular endothelial cells. METHODS We assessed endothelial activation in the presence or absence of ruxolitinib or fedratinib by using immunoblots, immunofluorescence, qRT-PCR, and function coagulation assays. Finally, we used endothelialized microfluidics perfused with blood from normal and JAK2V617F+ individuals to evaluate whether ruxolitinib and fedratinib changed cell adhesion. RESULTS We found that both ruxolitinib and fedratinib reduced endothelial cell phospho-STAT1 and STAT3 signaling and attenuated nuclear phospho-NK-κB and phospho-c-Jun localization. JAK-STAT inhibition also limited secretion of proadhesive and procoagulant P-selectin and von Willebrand factor and proinflammatory IL-6. Likewise, we found that JAK-STAT inhibition reduced endothelial tissue factor and urokinase plasminogen activator expression and activity. CONCLUSIONS By using endothelialized microfluidics perfused with whole blood samples, we demonstrated that endothelial treatment with JAK-STAT inhibitors prevented rolling of both healthy control and JAK2V617F MPN leukocytes. Together, these findings demonstrate that JAK-STAT inhibitors reduce the upregulation of critical prothrombotic pathways and prevent increased leukocyte-endothelial adhesion.
Collapse
Affiliation(s)
- Joan D Beckman
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA.
| | - Angelica DaSilva
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Elena Aronovich
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Aithanh Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Julia Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Geneva Hargis
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - David Reynolds
- Department of Biomedical Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gregory M Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brian Betts
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - David K Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
26
|
Grant AH, Rodriguez AC, Rodriguez Moncivais OJ, Sun S, Li L, Mohl JE, Leung MY, Kirken RA, Rodriguez G. JAK1 Pseudokinase V666G Mutant Dominantly Impairs JAK3 Phosphorylation and IL-2 Signaling. Int J Mol Sci 2023; 24:ijms24076805. [PMID: 37047778 PMCID: PMC10095075 DOI: 10.3390/ijms24076805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/17/2023] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
Overactive Janus kinases (JAKs) are known to drive leukemia, making them well-suited targets for treatment. We sought to identify new JAK-activating mutations and instead found a JAK1-inactivating pseudokinase mutation, V666G. In contrast to other pseudokinase mutations that canonically lead to an active kinase, the JAK1 V666G mutation led to under-activation seen by reduced phosphorylation. To understand the functional role of JAK1 V666G in modifying kinase activity we investigated its influence on other JAK kinases and within the Interleukin-2 pathway. JAK1 V666G not only inhibited its own activity, but its presence could inhibit other JAK kinases. These findings provide new insights into the potential of JAK1 pseudokinase to modulate its own activity, as well as of other JAK kinases. Thus, the features of the JAK1 V666 region in modifying JAK kinases can be exploited to allosterically inhibit overactive JAKs.
Collapse
Affiliation(s)
- Alice H. Grant
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Alejandro C. Rodriguez
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Omar J. Rodriguez Moncivais
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Shengjie Sun
- Department of Physics, The University of Texas at El Paso, El Paso, TX 79968, USA
- Computational Science Program, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Lin Li
- Department of Physics, The University of Texas at El Paso, El Paso, TX 79968, USA
- Computational Science Program, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Jonathon E. Mohl
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Mathematical Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Ming-Ying Leung
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
- Computational Science Program, The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Mathematical Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Robert A. Kirken
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Georgialina Rodriguez
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
27
|
Torres S, Ortiz C, Bachtler N, Gu W, Grünewald LD, Kraus N, Schierwagen R, Hieber C, Meier C, Tyc O, Joseph Brol M, Uschner FE, Nijmeijer B, Welsch C, Berres M, Garcia‐Ruiz C, Fernandez‐Checa JC, Trautwein C, Vogl TJ, Zeuzem S, Trebicka J, Klein S. Janus kinase 2 inhibition by pacritinib as potential therapeutic target for liver fibrosis. Hepatology 2023; 77:1228-1240. [PMID: 35993369 PMCID: PMC10026969 DOI: 10.1002/hep.32746] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Janus kinase 2 (JAK2) signaling is increased in human and experimental liver fibrosis with portal hypertension. JAK2 inhibitors, such as pacritinib, are already in advanced clinical development for other indications and might also be effective in liver fibrosis. Here, we investigated the antifibrotic role of the JAK2 inhibitor pacritinib on activated hepatic stellate cells (HSCs) in vitro and in two animal models of liver fibrosis in vivo . APPROACH AND RESULTS Transcriptome analyses of JAK2 in human livers and other targets of pacritinib have been shown to correlate with profibrotic factors. Although transcription of JAK2 correlated significantly with type I collagen expression and other profibrotic genes, no correlation was observed for interleukin-1 receptor-associated kinase and colony-stimulating factor 1 receptor. Pacritinib decreased gene expression of fibrosis markers in mouse primary and human-derived HSCs in vitro . Moreover, pacritinib decreased the proliferation, contraction, and migration of HSCs. C 57 BL/6J mice received ethanol in drinking water (16%) or Western diet in combination with carbon tetrachloride intoxication for 7 weeks to induce alcoholic or nonalcoholic fatty liver disease. Pacritinib significantly reduced liver fibrosis assessed by gene expression and Sirius red staining, as well as HSC activation assessed by alpha-smooth muscle actin immunostaining in fibrotic mice. Furthermore, pacritinib decreased the gene expression of hepatic steatosis markers in experimental alcoholic liver disease. Additionally, pacritinib protected against liver injury as assessed by aminotransferase levels. CONCLUSIONS This study demonstrates that the JAK2 inhibitor pacritinib may be promising for the treatment of alcoholic and nonalcoholic liver fibrosis and may be therefore relevant for human pathology.
Collapse
Affiliation(s)
- Sandra Torres
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Liver Unit‐IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
| | - Cristina Ortiz
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Nadine Bachtler
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Wenyi Gu
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Leon D. Grünewald
- Department of Diagnostic and Interventional Radiology, Universit+y Hospital Frankfurt, Frankfurt am Main, Germany
| | - Nico Kraus
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Robert Schierwagen
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Christoph Hieber
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Caroline Meier
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Olaf Tyc
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Maximilian Joseph Brol
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Frank Erhard Uschner
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Bart Nijmeijer
- Research and Development Department, Linxis BV, Amsterdam, The Netherlands
| | - Christoph Welsch
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Marie‐Luise Berres
- Department of Internal Medicine III, Aachen University Hospital, Aachen, Germany
| | - Carmen Garcia‐Ruiz
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Liver Unit‐IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
- Department of Medicine, University of Southern California, Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Jose Carlos Fernandez‐Checa
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Liver Unit‐IDIBAPS and Centro de Investigación Biomédica en Red (CIBERehd), Barcelona, Spain
- Department of Medicine, University of Southern California, Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Christian Trautwein
- Department of Internal Medicine III, Aachen University Hospital, Aachen, Germany
| | - Thomas J. Vogl
- Department of Diagnostic and Interventional Radiology, Universit+y Hospital Frankfurt, Frankfurt am Main, Germany
| | - Stefan Zeuzem
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
- European Foundation for the Study of Chronic Liver Failure – EF Clif, Barcelona, Spain
| | - Sabine Klein
- Department of Internal Medicine I, Goethe University Clinic Frankfurt, Frankfurt, Germany
- Department of Internal Medicine B, University of Münster, Münster, Germany
| |
Collapse
|
28
|
Mascarenhas J, Gleitz HFE, Chifotides HT, Harrison CN, Verstovsek S, Vannucchi AM, Rampal RK, Kiladjian JJ, Vainchenker W, Hoffman R, Schneider RK, List AF. Biological drivers of clinical phenotype in myelofibrosis. Leukemia 2023; 37:255-264. [PMID: 36434065 PMCID: PMC9898039 DOI: 10.1038/s41375-022-01767-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022]
Abstract
Myelofibrosis (MF) is a myeloproliferative disorder that exhibits considerable biological and clinical heterogeneity. At the two ends of the disease spectrum are the myelodepletive or cytopenic phenotype and the myeloproliferative phenotype. The cytopenic phenotype has a high prevalence in primary MF (PMF) and is characterized by low blood counts. The myeloproliferative phenotype is typically associated with secondary MF (SMF), mild anemia, minimal need for transfusion support, and normal to mild thrombocytopenia. Differences in somatic driver mutations and allelic burden, as well as the acquisition of non-driver mutations further influences these phenotypic differences, prognosis, and response to therapies such as JAK2 inhibitors. The outcome of patients with the cytopenic phenotype are comparatively worse and frequently pose a challenge to treat given the inherent exacerbation of cytopenias. Recent data indicate that an innate immune deregulated state that hinges on the myddosome-IRAK-NFκB axis favors the cytopenic myelofibrosis phenotype and offers opportunity for novel treatment approaches. We will review the biological and clinical features of the MF disease spectrum and associated treatment considerations.
Collapse
Affiliation(s)
- John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Hélène F E Gleitz
- Department of Developmental Biology, Erasmus Medical Center, Rotterdam, The Netherlands
- Oncode Institute, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Helen T Chifotides
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Srdan Verstovsek
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Raajit K Rampal
- Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | | | | | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rebekka K Schneider
- Department of Developmental Biology, Erasmus Medical Center, Rotterdam, The Netherlands
- Oncode Institute, Erasmus Medical Center, Rotterdam, The Netherlands
- Institute of Cell and Tumor Biology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | | |
Collapse
|
29
|
Mosale Seetharam S, Liu Y, Wu J, Fechter L, Murugesan K, Maecker H, Gotlib J, Zehnder J, Paulmurugan R, Krishnan A. Enkurin: A novel marker for myeloproliferative neoplasms from platelet, megakaryocyte, and whole blood specimens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.07.523111. [PMID: 36712071 PMCID: PMC9881897 DOI: 10.1101/2023.01.07.523111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Impaired protein homeostasis, though well established in age-related disorders, has been linked in recent research with the pathogenesis of myeloproliferative neoplasms (MPNs). As yet, however, little is known about MPN-specific modulators of proteostasis, thus impeding our ability for increased mechanistic understanding and discovery of additional therapeutic targets. Loss of proteostasis, in itself, is traced to dysregulated mechanisms in protein folding and intracellular calcium signaling at the endoplasmic reticulum (ER). Here, using ex vivo and in vitro systems (including CD34 + cultures from patient bone marrow, and healthy cord/peripheral blood specimens), we extend our prior data from MPN patient platelet RNA sequencing, and discover select proteostasis-associated markers at RNA and/or protein levels in each of platelets, parent megakaryocytes, and whole blood specimens. Importantly, we identify a novel role in MPNs for enkurin ( ENKUR ), a calcium mediator protein, implicated originally only in spermatogenesis. Our data reveal consistent ENKUR downregulation at both RNA and protein levels across MPN patient specimens and experimental models, with a concomitant upregulation of a cell cycle marker, CDC20 . Silencing of ENKUR by shRNA in CD34 + derived megakaryocytes further confirm this association with CDC20 at both RNA and protein levels; and indicate a likely role for the PI3K/Akt pathway. The inverse association of ENKUR and CDC20 expression was further confirmed upon treatment with thapsigargin (an agent that causes protein misfolding in the ER by selective loss of calcium) in both megakaryocyte and platelet fractions at RNA and protein levels. Together, our work sheds light on enkurin as a novel marker of MPN pathogenesis beyond the genetic alterations; and indicates further mechanistic investigation into a role for dysregulated calcium homeostasis, and ER and protein folding stress in MPN transformation. VISUAL ABSTRACT Key Points Enkurin, a calcium adaptor protein, is identified as a novel marker of pathogenesis in MPNs.MPN megakaryocyte and platelet expression of enkurin at RNA and protein levels is inversely associated with a cell differentiation cycle gene, CDC20.Likely role for dysregulated calcium homeostasis, and ER and protein folding stress in MPN transformation.
Collapse
Affiliation(s)
| | - Yi Liu
- Department of Radiology, Stanford University, Stanford, CA
| | - Jason Wu
- High-Throughput Bioscience Center (HTBC), Stanford University School of Medicine, Stanford, CA
| | - Lenn Fechter
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | | | - Holden Maecker
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA
| | - Jason Gotlib
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - James Zehnder
- Department of Pathology, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Anandi Krishnan
- Department of Pathology, Stanford University, Stanford, CA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
30
|
Sanachai K, Mahalapbutr P, Tabtimmai L, Seetaha S, Kaekratoke N, Chamni S, Azam SS, Choowongkomon K, Rungrotmongkol T. In Silico and In Vitro Study of Janus Kinases Inhibitors from Naphthoquinones. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020597. [PMID: 36677654 PMCID: PMC9866339 DOI: 10.3390/molecules28020597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023]
Abstract
Janus kinases (JAKs) are involved in numerous cellular signaling processes related to immune cell functions. JAK2 and JAK3 are associated with the pathogenesis of leukemia and common lymphoid-derived illnesses. JAK2/3 inhibitors could reduce the risk of various diseases by targeting this pathway. Herein, the naphthoquinones were experimentally and theoretically investigated to identify novel JAK2/3 inhibitors. Napabucasin and 2'-methyl napabucasin exhibited potent cell growth inhibition in TF1 (IC50 = 9.57 and 18.10 μM) and HEL (IC50 = 3.31 and 6.65 μM) erythroleukemia cell lines, and they significantly inhibited JAK2/3 kinase activity (in a nanomolar range) better than the known JAK inhibitor, tofacitinib. Flow cytometric analysis revealed that these two compounds induced apoptosis in TF1 cells in a time and dose-dependent manner. From the molecular dynamics study, both compounds formed hydrogen bonds with Y931 and L932 residues and hydrophobically contacted with the conserved hinge region, G loop, and catalytic loop of the JAK2. Our obtained results suggested that napabucasin and its methylated analog were potential candidates for further development of novel anticancer drug targeting JAKs.
Collapse
Affiliation(s)
- Kamonpan Sanachai
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panupong Mahalapbutr
- Department of Biochemistry, Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Lueacha Tabtimmai
- Department of Biotechnology, Faculty of Applied Science, King Mongkut’s University of Technology of North Bangkok, Bangkok 10800, Thailand
| | - Supaphorn Seetaha
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Nantawat Kaekratoke
- Department of Materials Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Supakarn Chamni
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Natural products and Nanoparticles Research Unit (NP2), Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Syed Sikander Azam
- Computational Biology Lab, National Center for Bioinformatics, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Correspondence: (K.C.); (T.R.); Tel.: +66-2-218-5426 (T.R.)
| | - Thanyada Rungrotmongkol
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (K.C.); (T.R.); Tel.: +66-2-218-5426 (T.R.)
| |
Collapse
|
31
|
Schmidts I, Haferlach T, Hoermann G. Precision Medicine in Therapy of Non-solid Cancer. Handb Exp Pharmacol 2023; 280:35-64. [PMID: 35989345 DOI: 10.1007/164_2022_608] [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] [Indexed: 06/15/2023]
Abstract
The development and approval of the tyrosine kinase inhibitor imatinib in 2001 has heralded the advance of directed therapy options. Today, an armamentarium of targeted therapeutics is available and enables the use of precision medicine in non-solid cancer. Precision medicine is guided by the detection of tumor-specific and targetable characteristics. These include pathogenic fusions and/or mutations, dependency on specific signaling pathways, and the expression of certain cell surface markers. Within the first part, we review approved targeted therapies for the compound classes of small molecule inhibitors, antibody-based therapies and cellular therapies. Particular consideration is given to the underlying pathobiology and the respective mechanism of action. The second part emphasizes on how biomarkers, whether they are of diagnostic, prognostic, or predictive relevance, are indispensable tools to guide therapy choice and management in precision medicine. Finally, the examples of acute myeloid leukemia, chronic lymphocytic leukemia, and chronic myeloid leukemia illustrate how integration of these biomarkers helps to tailor therapy.
Collapse
|
32
|
Sanachai K, Mahalapbutr P, Tabtimmai L, Seetaha S, Kittikool T, Yotphan S, Choowongkomon K, Rungrotmongkol T. Discovery of JAK2/3 Inhibitors from Quinoxalinone-Containing Compounds. ACS OMEGA 2022; 7:33587-33598. [PMID: 36157733 PMCID: PMC9494680 DOI: 10.1021/acsomega.2c04769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/26/2022] [Indexed: 06/16/2023]
Abstract
Janus kinases (JAKs) are involved in a wide variety of cell signaling associated with T-cell and B-cell mediated diseases. The pathogenesis of common lymphoid-derived diseases and leukemia cancer has been implicated in JAK2 and JAK3. Therefore, to decrease the risk of these diseases, targeting this pathway using JAK2/3 inhibitors could serve as a valuable research tool. Herein, we used a combination of the computational and biological approaches to identify the quinoxalinone-based dual inhibitors of JAK2/3. First, an in-house library of 49 quinoxalinones was screened by molecular docking. Then, the inhibitory activities of 17 screened compounds against both JAKs as well as against two human erythroleukemia cell lines, TF1 and HEL were examined. The obtained results revealed that several quinoxalinones could potentially inhibit JAK2/3, and among them, ST4j showed strong inhibition against JAKs with the IC50 values of 13.00 ± 1.31 nM for JAK2 and 14.86 ± 1.29 nM for JAK3, which are better than ruxolitinib and tofacitinib. In addition, ST4j potentially inhibited TF1 cells (IC50 of 15.53 ± 0.82 μM) and HEL cells (IC50 of 17.90 ± 1.36 μM), similar to both tofacitinib ruxolitinib. Mechanistically, ST4j inhibited JAK2 autophosphorylation and induced cell apoptosis in dose- and time-dependent manners. From molecular dynamics simulations, ST4j was mainly stabilized by van der Waals interactions, and its hydroxyl group could form hydrogen bonds in the hinge region at residues S936 and R938 of JAK2. This research highlights the potential of ST4j to be a novel therapeutic agent for the treatment of lymphoid-derived diseases and leukemia cancer.
Collapse
Affiliation(s)
- Kamonpan Sanachai
- Center
of Excellence in Structural and Computational Biology Research Unit,
Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panupong Mahalapbutr
- Department
of Biochemistry, and Center for Translational Medicine, Faculty of
Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Lueacha Tabtimmai
- Department
of Biotechnology, Faculty of Applied Science, King Mongkut’s University of Technology of North Bangkok, Bangkok 10800, Thailand
| | - Supaphorn Seetaha
- Department
of Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
| | - Tanakorn Kittikool
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| | - Sirilata Yotphan
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| | - Kiattawee Choowongkomon
- Department
of Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
| | - Thanyada Rungrotmongkol
- Center
of Excellence in Structural and Computational Biology Research Unit,
Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Program
in
Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
33
|
Sanachai K, Mahalapbutr P, Hengphasatporn K, Shigeta Y, Seetaha S, Tabtimmai L, Langer T, Wolschann P, Kittikool T, Yotphan S, Choowongkomon K, Rungrotmongkol T. Pharmacophore-Based Virtual Screening and Experimental Validation of Pyrazolone-Derived Inhibitors toward Janus Kinases. ACS OMEGA 2022; 7:33548-33559. [PMID: 36157769 PMCID: PMC9494641 DOI: 10.1021/acsomega.2c04535] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/26/2022] [Indexed: 06/16/2023]
Abstract
Janus kinases (JAKs) are nonreceptor protein tyrosine kinases that play a role in a broad range of cell signaling. JAK2 and JAK3 have been involved in the pathogenesis of common lymphoid-derived diseases and leukemia cancer. Thus, inhibition of both JAK2 and JAK3 can be a potent strategy to reduce the risk of these diseases. In the present study, the pharmacophore models built based on the commercial drug tofacitinib and the JAK2/3 proteins derived from molecular dynamics (MD) trajectories were employed to search for a dual potent JAK2/3 inhibitor by a pharmacophore-based virtual screening of 54 synthesized pyrazolone derivatives from an in-house data set. Twelve selected compounds from the virtual screening procedure were then tested for their inhibitory potency against both JAKs in the kinase assay. The in vitro kinase inhibition experiment indicated that compounds 3h, TK4g, and TK4b can inhibit both JAKs in the low nanomolar range. Among them, the compound TK4g showed the highest protein kinase inhibition with the half-maximal inhibitory concentration (IC50) value of 12.61 nM for JAK2 and 15.80 nM for JAK3. From the MD simulations study, it could be found that the sulfonamide group of TK4g can form hydrogen bonds in the hinge region at residues E930 and L932 of JAK2 and E903 and L905 of JAK3, while van der Waals interaction also plays a dominant role in ligand binding. Altogether, TK4g, found by virtual screening and biological tests, could serve as a novel therapeutical lead candidate.
Collapse
Affiliation(s)
- Kamonpan Sanachai
- Center
of Excellence in Structural and Computational Biology Research Unit,
Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok10330, Thailand
| | - Panupong Mahalapbutr
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen40002, Thailand
| | - Kowit Hengphasatporn
- Center
for Computational Sciences, University of
Tsukuba, 1-1-1 Tennodai, Tsukuba305-8577, Ibaraki, Japan
| | - Yasuteru Shigeta
- Center
for Computational Sciences, University of
Tsukuba, 1-1-1 Tennodai, Tsukuba305-8577, Ibaraki, Japan
| | - Supaphorn Seetaha
- Department
of Biochemistry, Faculty of Science, Kasetsart
University, Bangkok10900, Thailand
| | - Lueacha Tabtimmai
- Department
of Biotechnology, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok10800, Thailand
| | - Thierry Langer
- Department
of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstraße 14, ViennaA-1090, Austria
| | - Peter Wolschann
- Institute
of Theoretical Chemistry, University of
Vienna, Vienna1090, Austria
| | - Tanakorn Kittikool
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Mahidol University, Rama VI Road, Bangkok10400, Thailand
| | - Sirilata Yotphan
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Mahidol University, Rama VI Road, Bangkok10400, Thailand
| | - Kiattawee Choowongkomon
- Department
of Biochemistry, Faculty of Science, Kasetsart
University, Bangkok10900, Thailand
| | - Thanyada Rungrotmongkol
- Center
of Excellence in Structural and Computational Biology Research Unit,
Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok10330, Thailand
- Program
in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok10330, Thailand
| |
Collapse
|
34
|
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: 10.3] [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
|
35
|
Downes CEJ, McClure BJ, McDougal DP, Heatley SL, Bruning JB, Thomas D, Yeung DT, White DL. JAK2 Alterations in Acute Lymphoblastic Leukemia: Molecular Insights for Superior Precision Medicine Strategies. Front Cell Dev Biol 2022; 10:942053. [PMID: 35903543 PMCID: PMC9315936 DOI: 10.3389/fcell.2022.942053] [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: 05/12/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer, arising from immature lymphocytes that show uncontrolled proliferation and arrested differentiation. Genomic alterations affecting Janus kinase 2 (JAK2) correlate with some of the poorest outcomes within the Philadelphia-like subtype of ALL. Given the success of kinase inhibitors in the treatment of chronic myeloid leukemia, the discovery of activating JAK2 point mutations and JAK2 fusion genes in ALL, was a breakthrough for potential targeted therapies. However, the molecular mechanisms by which these alterations activate JAK2 and promote downstream signaling is poorly understood. Furthermore, as clinical data regarding the limitations of approved JAK inhibitors in myeloproliferative disorders matures, there is a growing awareness of the need for alternative precision medicine approaches for specific JAK2 lesions. This review focuses on the molecular mechanisms behind ALL-associated JAK2 mutations and JAK2 fusion genes, known and potential causes of JAK-inhibitor resistance, and how JAK2 alterations could be targeted using alternative and novel rationally designed therapies to guide precision medicine approaches for these high-risk subtypes of ALL.
Collapse
Affiliation(s)
- Charlotte EJ. Downes
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Barbara J. McClure
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Daniel P. McDougal
- School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, SA, Australia
| | - Susan L. Heatley
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Australian and New Zealand Children’s Oncology Group (ANZCHOG), Clayton, VIC, Australia
| | - John B. Bruning
- School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, SA, Australia
| | - Daniel Thomas
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - David T. Yeung
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Department of Haematology, Royal Adelaide Hospital and SA Pathology, Adelaide, SA, Australia
| | - Deborah L. White
- Blood Cancer Program, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Australian and New Zealand Children’s Oncology Group (ANZCHOG), Clayton, VIC, Australia
| |
Collapse
|
36
|
Ibarra J, Elbanna YA, Kurylowicz K, Ciboddo M, Greenbaum HS, Arellano NS, Rodriguez D, Evers M, Bock-Hughes A, Liu C, Smith Q, Lutze J, Baumeister J, Kalmer M, Olschok K, Nicholson B, Silva D, Maxwell L, Dowgielewicz J, Rumi E, Pietra D, Casetti IC, Catricala S, Koschmieder S, Gurbuxani S, Schneider RK, Oakes SA, Elf SE. Type I but Not Type II Calreticulin Mutations Activate the IRE1α/XBP1 Pathway of the Unfolded Protein Response to Drive Myeloproliferative Neoplasms. Blood Cancer Discov 2022; 3:298-315. [PMID: 35405004 PMCID: PMC9338758 DOI: 10.1158/2643-3230.bcd-21-0144] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/21/2022] [Accepted: 04/08/2022] [Indexed: 01/09/2023] Open
Abstract
Approximately 20% of patients with myeloproliferative neoplasms (MPN) harbor mutations in the gene calreticulin (CALR), with 80% of those mutations classified as either type I or type II. While type II CALR-mutant proteins retain many of the Ca2+ binding sites present in the wild-type protein, type I CALR-mutant proteins lose these residues. The functional consequences of this differential loss of Ca2+ binding sites remain unexplored. Here, we show that the loss of Ca2+ binding residues in the type I mutant CALR protein directly impairs its Ca2+ binding ability, which in turn leads to depleted endoplasmic reticulum (ER) Ca2+ and subsequent activation of the IRE1α/XBP1 pathway of the unfolded protein response. Genetic or pharmacologic inhibition of IRE1α/XBP1 signaling induces cell death in type I mutant but not type II mutant or wild-type CALR-expressing cells, and abrogates type I mutant CALR-driven MPN disease progression in vivo. SIGNIFICANCE Current targeted therapies for CALR-mutated MPNs are not curative and fail to differentiate between type I- versus type II-driven disease. To improve treatment strategies, it is critical to identify CALR mutation type-specific vulnerabilities. Here we show that IRE1α/XBP1 represents a unique, targetable dependency specific to type I CALR-mutated MPNs. This article is highlighted in the In This Issue feature, p. 265.
Collapse
Affiliation(s)
- Juan Ibarra
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Yassmin A. Elbanna
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Katarzyna Kurylowicz
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Michele Ciboddo
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Harrison S. Greenbaum
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Nicole S. Arellano
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Deborah Rodriguez
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Maria Evers
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Althea Bock-Hughes
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Chenyu Liu
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Quinn Smith
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Julian Lutze
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois
| | - Julian Baumeister
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Milena Kalmer
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Kathrin Olschok
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Benjamin Nicholson
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Diane Silva
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Luke Maxwell
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Jonathan Dowgielewicz
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Elisa Rumi
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Daniela Pietra
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | | | - Silvia Catricala
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | | | - Rebekka K. Schneider
- Department of Cell Biology, Institute for Biomedical Technologies, RWTH Aachen University, Aachen, Germany
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Scott A. Oakes
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Shannon E. Elf
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| |
Collapse
|
37
|
Siriwaseree J, Sanachai K, Aiebchun T, Tabtimmai L, Kuaprasert B, Choowongkomon K. Synchrotron Fourier Transform Infrared Microscopy Spectra in Cellular Effects of Janus Kinase Inhibitors on Myelofibrosis Cancer Cells. ACS OMEGA 2022; 7:22797-22803. [PMID: 35811912 PMCID: PMC9260937 DOI: 10.1021/acsomega.2c02404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Janus kinase (JAK) deregulation of the JAK/signal transducers and activators of transcription pathway leads to myelofibrosis that can be treated by JAK inhibitors including Ruxolitinib and Tofacitinib. Even though both inhibitors are effective against myelofibrosis, each of them has a different mode of action in the cells. Ruxolitinib is an inhibitor for selective JAK1/2, and Tofacitinib is an inhibitor for JAK3. This study evaluated the chemical fingerprints of TF-1 cells after JAK inhibitor treatments by the synchrotron Fourier transform infrared microspectroscopy (S-FTIR) spectrum. Tofacitinib and Ruxolitinib treatments in TF-1 cells were applied with a chemical fingerprint approach in S-FTIR spectroscopy and in vitro cytotoxicity in a cell-based assay. Principal component analysis or PCA was utilized to classify three cell treatments with three biochemical alteration absorbances of lipid vibration by the C-H stretching, protein amide I that appeared from the C=O stretching, and a P=O phosphodiester bond from nucleic acids. The results showed that the inhibition effect of Ruxolitinib on the TF-1 cell lines was two-fold higher than Tofacitinib. PCA distinguishes untreated and drug-treated cells by detecting cellular biochemical alteration. The loading plots identify that proteins and nucleic acids were the different main components in disparate cell treatments. Tofacitinib was distinct from the others in lipid and nucleic acid. The second derivative spectra of the three molecular components had decreased lipid production and accumulation, changes in secondary structures in proteins, and a high level of RNA overexpression in cell treatment. The JAK inhibitors caused different spectroscopic biomarkers of the modifications of secondary protein conformation, stimulated cell lipid accumulation, and phosphorylation from untreated cells. The alteration of cellular biochemical components suggests that FTIR is a potential tool to analyze specific patterns of drug cellular responses at the molecular level.
Collapse
Affiliation(s)
- Jeeraprapa Siriwaseree
- Faculty
of Science, Department of Biochemistry, Kasetsart University, Bangkok 10900, Thailand
| | - Kamonpan Sanachai
- Faculty
of Science, Department of Biochemistry, Structural and Computational
Biology Research Unit, Chulalongkorn University, Bangkok 10330, Thailand
| | - Thitinan Aiebchun
- Faculty
of Science, Department of Biochemistry, Kasetsart University, Bangkok 10900, Thailand
| | - Lueacha Tabtimmai
- Faculty
of Applied Science, Department of Biotechnology, King Mongkut’s University of Technology of North Bangkok, Bangkok 10800, Thailand
| | - Buabarn Kuaprasert
- Synchrotron
Light Research Institute (Public Organization), Nakhon Ratchasrima 30000, Thailand
| | - Kiattawee Choowongkomon
- Faculty
of Science, Department of Biochemistry, Kasetsart University, Bangkok 10900, Thailand
| |
Collapse
|
38
|
Salter B, Burns I, Fuller K, Eshaghpour A, Lionel AC, Crowther M. Tyrosine kinase inhibitors and tumor lysis syndrome in hematologic malignancies: A systemic review. Eur J Haematol 2022; 109:166-181. [PMID: 35531791 DOI: 10.1111/ejh.13786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Effective treatments for hematologic malignancies include therapies that target tyrosine kinase (TK) signaling pathways. Tumor lysis syndrome (TLS) is an oncologic emergency that can occur due to rapid turnover following the initiation of treatments for hematologic malignancy. The incidence of TLS is under-reported and it is unclear as to whether TK inhibitors (TKIs) are associated with TLS. OBJECTIVE To conduct a systematic review to determine the incidence of TLS with TKIs. METHODS A search was performed using EMBASE, MEDLINE, and Web of Science electronic databases, as well as a manual search of the American Society of Hematology and American Society of Clinical Oncology abstract databases. Keywords included: "tumor lysis syndrome," "tyrosine kinase inhibitors," "lymphoma," and "leukemia." RESULTS We identified a total of 57 publications that commented on the incidence of TLS with TKIs for hematologic malignancy. Thirty-nine of those publications reported TLS as an adverse event. TLS was described as an adverse event among essentially all the subclasses of TKIs that are used to manage hematologic malignancies. CONCLUSION The overall number of articles commenting on TLS as an adverse event is sparse and there needs to be more transparency regarding the incidence of TLS when employing newer targeted therapies. Physicians should consider the risk of TLS on an individual basis and the added risk of TLS when using TKIs to treat hematologic malignancy.
Collapse
Affiliation(s)
| | - Ian Burns
- McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | |
Collapse
|
39
|
A Comprehensive Overview of Globally Approved JAK Inhibitors. Pharmaceutics 2022; 14:pharmaceutics14051001. [PMID: 35631587 PMCID: PMC9146299 DOI: 10.3390/pharmaceutics14051001] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [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
|
40
|
JAK2 Inhibitor, Fedratinib, Inhibits P-gp Activity and Co-Treatment Induces Cytotoxicity in Antimitotic Drug-Treated P-gp Overexpressing Resistant KBV20C Cancer Cells. Int J Mol Sci 2022; 23:ijms23094597. [PMID: 35562984 PMCID: PMC9100550 DOI: 10.3390/ijms23094597] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
P-glycoprotein (P-gp) overexpression is one of the major mechanisms of multidrug resistance (MDR). Previously, co-treatment with Janus kinase 2 (JAK2) inhibitors sensitized P-gp-overexpressing drug-resistant cancer cells. In this study, we assessed the cytotoxic effects of JAK2 inhibitor, fedratinib, on drug-resistant KBV20C cancer cells. We found that co-treatment with fedratinib at low doses induced cytotoxicity in KBV20C cells treated with vincristine (VIC). However, fedratinib-induced cytotoxicity was little effect on VIC-treated sensitive KB parent cells, suggesting that these effects are specific to resistant cancer cells. Fluorescence-activated cell sorting (FACS), Western blotting, and annexin V analyses were used to further investigate fedratinib’s mechanism of action in VIC-treated KBV20C cells. We found that fedratinib reduced cell viability, increased G2 arrest, and upregulated apoptosis when used as a co-treatment with VIC. G2 phase arrest and apoptosis in VIC–fedratinib-co-treated cells resulted from the upregulation of p21 and the DNA damaging marker pH2AX. Compared with dimethyl sulfoxide (DMSO)-treated cells, fedratinib-treated KBV20C cells showed two-fold higher P-gp-inhibitory activity, indicating that VIC–fedratinib sensitization is dependent on the activity of fedratinib. Similar to VIC, fedratinib co-treatment with other antimitotic drugs (i.e., eribulin, vinorelbine, and vinblastine) showed increased cytotoxicity in KBV20C cells. Furthermore, VIC–fedratinib had similar cytotoxic effects to co-treatment with other JAK2 inhibitors (i.e., VIC–CEP-33779 or VIC–NVP-BSK805) at the same dose; similar cytotoxic mechanisms (i.e., early apoptosis) were observed between treatments, suggesting that co-treatment with JAK2 inhibitors is generally cytotoxic to P-gp-overexpressing resistant cancer cells. Given that fedratinib is FDA-approved, our findings support its application in the co-treatment of P-gp-overexpressing cancer patients showing MDR.
Collapse
|
41
|
Li Y, Wei L, Meinsohn MC, Suliman R, Chauvin M, Berstler J, Hartland K, Jensen MM, Sicher NA, Nagykery N, Donahoe PK, Pepin D. A screen of repurposed drugs identifies AMHR2/MISR2 agonists as potential contraceptives. Proc Natl Acad Sci U S A 2022; 119:e2122512119. [PMID: 35380904 PMCID: PMC9169708 DOI: 10.1073/pnas.2122512119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 11/18/2022] Open
Abstract
We identified the anti-Mullerian hormone (also known as Müllerian inhibiting substance or MIS) as an inhibitory hormone that induces long-term contraception in mammals. The type II receptor to this hormone, AMHR2 (also known as MISR2), represents a promising druggable target for the modulation of female reproduction with a mechanism of action distinct from steroidal contraceptives. We designed an in vitro platform to screen and validate small molecules that can activate MISR2 signaling and suppress ovarian folliculogenesis. Using a bone morphogenesis protein (BMP)–response element luciferase reporter cell–based assay, we screened 5,440 compounds from a repurposed drug library. Positive hits in this screen were tested for specificity and potency in luciferase dose–response assays, and biological activity was tested in ex vivo Mullerian duct regression bioassays. Selected candidates were further evaluated in ex vivo follicle/ovary culture assays and in vivo in mice and rats. Here, we report that SP600125, CYC-116, gandotinib, and ruxolitinib can specifically inhibit primordial follicle activation and repress folliculogenesis by stimulating the MISR2 pathway.
Collapse
Affiliation(s)
- Yi Li
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Lina Wei
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Marie-Charlotte Meinsohn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Rana Suliman
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Maeva Chauvin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Jim Berstler
- Center for the Development of Therapeutics (CDoT), Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142
| | - Kate Hartland
- Center for the Development of Therapeutics (CDoT), Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142
| | - Mark M Jensen
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Natalie A Sicher
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Nicholas Nagykery
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - David Pepin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02114
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
42
|
Loscocco GG, Vannucchi AM. Role of JAK inhibitors in myeloproliferative neoplasms: current point of view and perspectives. Int J Hematol 2022; 115:626-644. [PMID: 35352288 DOI: 10.1007/s12185-022-03335-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/06/2022] [Accepted: 03/15/2022] [Indexed: 12/29/2022]
Abstract
Classic Philadelphia-negative myeloproliferative neoplasms (MPN) include polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF), classified as primary (PMF), or secondary to PV or ET. All MPN, regardless of the underlying driver mutation in JAK2/CALR/MPL, are invariably associated with dysregulation of JAK/STAT pathway. The discovery of JAK2V617F point mutation prompted the development of small molecules inhibitors of JAK tyrosine kinases (JAK inhibitors-JAKi). To date, among JAKi, ruxolitinib (RUX) and fedratinib (FEDR) are approved for intermediate and high-risk MF, and RUX is also an option for high-risk PV patients inadequately controlled by or intolerant to hydroxyurea. While not yet registered, pacritinib (PAC) and momelotinib (MMB), proved to be effective particularly in thrombocytopenic and anemic MF patients, respectively. In most cases, JAKi are effective in reducing splenomegaly and alleviating disease-related symptoms. However, almost 50% lose response by three years and dose-dependent toxicities may lead to suboptimal dosing or treatment discontinuation. To date, although not being disease-modifying agents, JAKi represent the therapeutic backbone particularly in MF patient. To optimize therapeutic strategies, many trials with drug combinations of JAKi with novel molecules are ongoing. This review critically discusses the role of JAKi in the modern management of patients with MPN.
Collapse
Affiliation(s)
- Giuseppe G Loscocco
- Department of Experimental and Clinical Medicine, University of Florence, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3 pad 27B, 50134, Florence, Italy
- Doctorate School GenOMec, University of Siena, Siena, Italy
| | - Alessandro M Vannucchi
- Department of Experimental and Clinical Medicine, University of Florence, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3 pad 27B, 50134, Florence, Italy.
| |
Collapse
|
43
|
Rah B, Rather RA, Bhat GR, Baba AB, Mushtaq I, Farooq M, Yousuf T, Dar SB, Parveen S, Hassan R, Mohammad F, Qassim I, Bhat A, Ali S, Zargar MH, Afroze D. JAK/STAT Signaling: Molecular Targets, Therapeutic Opportunities, and Limitations of Targeted Inhibitions in Solid Malignancies. Front Pharmacol 2022; 13:821344. [PMID: 35401182 PMCID: PMC8987160 DOI: 10.3389/fphar.2022.821344] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/14/2022] [Indexed: 12/24/2022] Open
Abstract
JAK/STAT signaling pathway is one of the important regulatory signaling cascades for the myriad of cellular processes initiated by various types of ligands such as growth factors, hormones, and cytokines. The physiological processes regulated by JAK/STAT signaling are immune regulation, cell proliferation, cell survival, apoptosis and hematopoiesis of myeloid and non-myeloid cells. Dysregulation of JAK/STAT signaling is reported in various immunological disorders, hematological and other solid malignancies through various oncogenic activation mutations in receptors, downstream mediators, and associated transcriptional factors such as STATs. STATs typically have a dual role when explored in the context of cancer. While several members of the STAT family are involved in malignancies, however, a few members which include STAT3 and STAT5 are linked to tumor initiation and progression. Other STAT members such as STAT1 and STAT2 are pivotal for antitumor defense and maintenance of an effective and long-term immune response through evolutionarily conserved programs. The effects of JAK/STAT signaling and the persistent activation of STATs in tumor cell survival; proliferation and invasion have made the JAK/STAT pathway an ideal target for drug development and cancer therapy. Therefore, understanding the intricate JAK/STAT signaling in the pathogenesis of solid malignancies needs extensive research. A better understanding of the functionally redundant roles of JAKs and STATs may provide a rationale for improving existing cancer therapies which have deleterious effects on normal cells and to identifying novel targets for therapeutic intervention in solid malignancies.
Collapse
|
44
|
Cooray S, Sabanathan S, Hacohen Y, Worth A, Eleftheriou D, Hemingway C. Treatment Strategies for Central Nervous System Effects in Primary and Secondary Haemophagocytic Lymphohistiocytosis in Children. Curr Treat Options Neurol 2022. [DOI: 10.1007/s11940-022-00705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Abstract
Purpose of Review
This review presents an appraisal of current therapeutic options for the treatment of central nervous system haemophagocytic lymphohistiocytosis (CNS-HLH) in the context of systemic disease, as well as when CNS features occur in isolation. We present the reader with a diagnostic approach to CNS-HLH and commonly used treatment protocols. We discuss and evaluate newer treatments on the horizon.
Recent Findings
Mortality is high in patients who do not undergo HSCT, and while larger studies are required to establish benefit in many treatments, a number of new treatments are currently being evaluated. Alemtuzumab is being used as a first-line treatment for CNS-HLH in a phase I/II multicentre prospective clinical trial as an alternative to traditional HLH-1994 and 2004 protocols. It has also been used successfully as a second-line agent for the treatment of isolated CNS-HLH that is refractory to standard treatment. Ruxolitinib and emapalumab are new immunotherapies that block the Janus kinase—Signal Transducer and Activator of Transcription (JAK-STAT) pathway that have shown efficacy in refractory HLH, including for CNS-HLH disease.
Summary
Treatment of CNS-HLH often requires HLH-94 or 2004 protocols followed by haematopoietic stem cell transplantation (HSCT) to maintain remission, although relapse can occur, particularly with reduced intensity conditioning if donor chimerism falls. CNS features have been shown to improve or stabilise following HSCT in CNS-HLH in the context of systemic disease and in isolated CNS-HLH. Encouraging reports of early cohort studies suggest alemtuzumab and the Janus kinase (JAK) inhibitor ruxolitinib offer potential salvage therapy for relapsed and refractory CNS-HLH. Newer immunotherapies such as tocilizumab and natalizumab have been shown to be beneficial in sporadic cases. CNS-HLH due to primary gene defects may be amenable to gene therapy in the future.
Collapse
|
45
|
Bochicchio MT, Di Battista V, Poggio P, Carrà G, Morotti A, Brancaccio M, Lucchesi A. Understanding Aberrant Signaling to Elude Therapy Escape Mechanisms in Myeloproliferative Neoplasms. Cancers (Basel) 2022; 14:cancers14040972. [PMID: 35205715 PMCID: PMC8870427 DOI: 10.3390/cancers14040972] [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: 12/28/2021] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/01/2023] Open
Abstract
Aberrant signaling in myeloproliferative neoplasms may arise from alterations in genes coding for signal transduction proteins or epigenetic regulators. Both mutated and normal cells cooperate, altering fragile balances in bone marrow niches and fueling persistent inflammation through paracrine or systemic signals. Despite the hopes placed in targeted therapies, myeloid proliferative neoplasms remain incurable diseases in patients not eligible for stem cell transplantation. Due to the emergence of drug resistance, patient management is often very difficult in the long term. Unexpected connections among signal transduction pathways highlighted in neoplastic cells suggest new strategies to overcome neoplastic cell adaptation.
Collapse
Affiliation(s)
- Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Valeria Di Battista
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Pietro Poggio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy;
| | - Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| | - Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
- Correspondence: (A.M.); (M.B.); (A.L.)
| |
Collapse
|
46
|
Exploring the oncogenic and therapeutic target potential of the MYB-TYK2 fusion gene in B-cell acute lymphoblastic leukemia. Cancer Gene Ther 2022; 29:1140-1152. [PMID: 35022522 DOI: 10.1038/s41417-021-00421-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/01/2021] [Accepted: 12/15/2021] [Indexed: 11/08/2022]
Abstract
TYK2-rearrangements have recently been identified in high-risk acute lymphoblastic leukemia (HR-ALL) cases and are associated with poor outcome. Current understanding of the leukemogenic potential and therapeutic targetability of activating TYK2 alterations in the ALL setting is unclear, thus further investigations are warranted. Consequently, we developed in vitro, and for the first time, in vivo models of B-cell ALL from a patient harboring the MYB-TYK2 fusion gene. These models revealed JAK/STAT signaling activation and the oncogenic potential of the MYB-TYK2 fusion gene in isolation. High throughput screening identified the HDAC inhibitor, vorinostat and the HSP90 inhibitor, tanespimycin plus the JAK inhibitor, cerdulatinib as the most effective agents against cells expressing the MYB-TYK2 alteration. Evaluation of vorinostat and cerdulatinib in pre-clinical models of MYB-TYK2-rearranged ALL demonstrated that both drugs exhibited anti-leukemic effects and reduced the disease burden in treated mice. Importantly, these findings indicate that activating TYK2 alterations can function as driver oncogenes rather than passenger or secondary events in disease development. In addition, our data provide evidence for use of vorinostat and cerdulatinib in the treatment regimens of patients with this rare yet aggressive type of high-risk ALL that warrants further investigation in the clinical setting.
Collapse
|
47
|
Abstract
Background This study analyses treatment patterns, health care resource utilization (HCRU), and costs in patients with myelofibrosis (MF) and a subgroup treated with ruxolitinib (RUX). Materials and Methods Treatment patterns, all-cause and MF-related HCRU, and costs were analyzed in adults with MF with continuous enrollment in a commercial or the Medicare Advantage health plan in the pre-index period, defined as the 12 months immediately prior to the index date (date of primary or secondary MF diagnosis), and the post-index period, defined as ≥6 months following the index date. In a subgroup analysis, outcomes were analyzed in patients treated with optimal RUX (OPT RUX, ≥30 mg) and suboptimal RUX (SUB RUX, <30 mg) in the pre-index RUX period, defined as the 3 months immediately prior to the index RUX date (first date for an RUX claim), and the post-index RUX period, defined as ≥6 months following the index RUX date. Results Of 2830 patients with an MF diagnosis, 1191 met eligibility requirements. The median age of patients was 72 years, 54% were male, and comorbidities were frequent. Sixty percent of patients received ≥1 line of therapy (LOT), of which 46% (n = 331) had ≥2 LOTs during the post-index MF period. Costs increased considerably 6-month pre-index to 6-month post-index (all-cause: cause ($24,216 to $48,966) and MF-related ($16,502 to $39,383), driven by inpatient stays and pharmacy costs. In the subgroup analysis, patients treated with RUX (n = 495) experienced significant disease burden and high costs, regardless of dose. A shorter duration of therapy and a higher rate of discontinuation were observed in patients treated with SUB RUX (n = 191) versus OPT RUX (n = 304). Conclusion These findings suggest a significant disease and economic impacts associated with MF patients that persists with RUX therapy, highlighting the need for additional therapeutic options for MF.
Collapse
Affiliation(s)
- Ronda Copher
- Corresponding author: Ronda Copher WW HEOR, US Hematology, Bristol-Myers Squibb, 100 Nassau Park Boulevard #300, Princeton, NJ 08540, USA; Tel:+1 908 673 9594;
| | | | - Aaron Gerds
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
48
|
Zeiser R, Socié G, Schroeder MA, Abhyankar S, Vaz CP, Kwon M, Clausen J, Volodin L, Giebel S, Chacon MJ, Meyers G, Ghosh M, Deeren D, Sanz J, Morariu-Zamfir R, Arbushites M, Lakshminarayanan M, Barbour AM, Chen YB. Efficacy and safety of itacitinib versus placebo in combination with corticosteroids for initial treatment of acute graft-versus-host disease (GRAVITAS-301): a randomised, multicentre, double-blind, phase 3 trial. Lancet Haematol 2022; 9:e14-e25. [DOI: 10.1016/s2352-3026(21)00367-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/21/2022]
|
49
|
Thomas JW, Jamy O, Shah MV, Vachhani P, Go RS, Goyal G. Risk of mortality and second malignancies in primary myelofibrosis before and after ruxolitinib approval. Leuk Res 2021; 112:106770. [PMID: 34920340 DOI: 10.1016/j.leukres.2021.106770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/02/2021] [Accepted: 12/04/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND Primary myelofibrosis (PMF) is associated with morbidity and mortality. Ruxolitinib gained US FDA approval for treatment of intermediate/high-risk PMF in November 2011. We evaluated differences in survival and second primary malignancy (SPM) incidence among US PMF patients in the years before and after ruxolitinib approval. METHODS We conducted a retrospective study utilizing the National Cancer Institute's Surveillance, Epidemiology, and End Results (SEER)-18 database for PMF patients. We divided patients into five-year cohorts pre- (2007-2011) and post-ruxolitinib (2012-2016) approval and compared relative survival rates (RSRs) to the standard population and standardized incidence rates (SIRs) of SPMs between cohorts. RESULTS We included 2020 patients diagnosed with PMF from 2007-2016 in this study. There was no difference in the four-year RSRs between cohorts (54 % vs. 57 %, p = 0.776). More patients developed SPMs in the post-ruxolitinib cohort (8% vs. 6%, p = 0.041). The majority of SPMs were hematologic with higher incidence of AML transformation in the post-ruxolitinib cohort (SIR 125.29 vs. 70.55). CONCLUSIONS PMF prognosis remains poor in the years following ruxolitinib's approval. SPM incidence including AML transformation is higher in the years after approval. Further studies are needed to determine the true impact of ruxolitnib on population outcomes.
Collapse
Affiliation(s)
- John W Thomas
- Tinsley Harrison Internal Medicine Residency Program, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Omer Jamy
- Division of Hematology and Oncology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
| | | | - Pankit Vachhani
- Division of Hematology and Oncology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Ronald S Go
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Gaurav Goyal
- Division of Hematology and Oncology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
| |
Collapse
|
50
|
Cheng YY, Yang X, Gao X, Song SX, Yang MF, Xie FM. LGR6 promotes glioblastoma malignancy and chemoresistance by activating the Akt signaling pathway. Exp Ther Med 2021; 22:1364. [PMID: 34659510 PMCID: PMC8515564 DOI: 10.3892/etm.2021.10798] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Chemoresistance is the primary cause of the poor outcome of glioblastoma multiforme (GBM) therapy. Leucine-rich repeat-containing G-protein coupled receptor 6 (LGR6) is involved in the growth and proliferation of several types of cancer, including gastric cancer and ovarian cancer. Therefore, the aim of the present study was to investigate the role of LGR6 in GBM malignancy and chemoresistance. Cell counting kit-8 and Matrigel®-Transwell assays were conducted to assess GBM cell viability and invasion. The effect of LGR6 on cell cycle progression and activation of Akt signaling was analyzed by performing propidium iodide staining and western blotting, respectively. The results demonstrated that LGR6, a microRNA-1236-3p target candidate, promoted GBM cell viability and invasion, and mediated temozolomide sensitivity in SHG-44 and U251 GBM cells. In addition, LGR6 triggered the activation of the Akt signaling pathway during GBM progression. Collectively, the results of the present study suggested that LGR6 promoted GBM malignancy and chemoresistance, at least in part, by activating the Akt signaling pathway. The results may aid with the identification of a novel therapeutic target and strategy for GBM.
Collapse
Affiliation(s)
- Yuan Yuan Cheng
- Department of Oncology, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Xue Yang
- Department of Oncology, Tai'an Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Xin Gao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, P.R. China
| | - Si Xin Song
- Department of Neurosurgery, The Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong 270000, P.R. China
| | - Ming Feng Yang
- Institute of Basic Medicine of Shangdong, First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong 270000, P.R. China
| | - Fang Min Xie
- Department of Neurosurgery, The Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, Shandong 270000, P.R. China
| |
Collapse
|