1
|
Herqash RN, Alkathiri FA, Darwish IA. Assessing pharmacokinetics and drug-drug interactions of the combination therapy of myelofibrosis with ruxolitinib and lenalidomide by a new eco-friendly HPLC method for their simultaneous determination in plasma. Cancer Chemother Pharmacol 2024; 94:747-761. [PMID: 39259291 DOI: 10.1007/s00280-024-04715-y] [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: 07/10/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
Ruxolitinib (RUX), a Janus kinase 2 (JAK2) inhibitor, and lenalidomide (LEN), an immunomodulatory agent, have recently been proposed as a combined treatment for myelofibrosis (MF). This combination has demonstrated improved efficacy, safety, and tolerability compared to monotherapy. To further refine these findings, an efficient analytical tool is needed to simultaneously determine RUX and LEN concentrations in blood plasma. This tool would enable the study of their pharmacokinetics, drug-drug interactions, and therapeutic monitoring during MF therapy. Unfortunately, such a method has not been existed in the literature. This study presents the first HPLC method with UV detection for the simultaneous quantitation of RUX and LEN in plasma. The method was validated according to the ICH guidelines for bioanalytical method validation. It exhibited linearity in the concentration ranges of 10 to 3150 ng mL- 1 for RUX and 80 to 5200 ng mL- 1 for LEN. The limits of quantitation were determined to be 25 and 90 ng mL- 1 for RUX and LEN, respectively. All other validation parameters were satisfactory. The HPLC-UV method was successfully employed to study the pharmacokinetics and drug-drug interactions of RUX and LEN in rats following oral administration of single doses. The results demonstrated that the pharmacokinetics of both drugs were changed substantially by their coadministration. LEN exhibited synergistic effects on the maximum plasma concentration (Cmax) and total bioavailability of RUX, meanwhile it exhibited diminishing effect on the values of volume of distribution (Vd) and clearance (CL). Additionally, RUX decreased the Cmax and total bioavailability of LEN, meanwhile it increased its Vd and CL. These data suggest that the use of RUX, as a combination with LEN, is a better therapeutic approach for MF, compared with RUX as a monotherapy. The effects of LEN on the pharmacokinetics of RUX should be considered and can be useful in determining the appropriate RUX dosage and dosing regimen to achieve the desired therapeutic effect when used as a combination therapy with LEN. The method's environmental friendliness was confirmed through three comprehensive tools. This method represents a valuable tool for determining the appropriate dosage and dosing regimen of RUX in combination therapy with LEN to achieve the desired therapeutic effect. Furthermore, it can aid in predicting drug distribution in different patients and assessing the drug accumulation or insufficient drug levels in specific body compartments.
Collapse
Affiliation(s)
- Rashed N Herqash
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Fai A Alkathiri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ibrahim A Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia.
| |
Collapse
|
2
|
Liu X, Wang B, Liu Y, Yu Y, Wan Y, Wu J, Wang Y. JAK2 inhibitors for the treatment of Philadelphia-negative myeloproliferative neoplasms: current status and future directions. Mol Divers 2024; 28:3445-3456. [PMID: 38006563 DOI: 10.1007/s11030-023-10742-3] [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: 07/23/2023] [Accepted: 10/05/2023] [Indexed: 11/27/2023]
Abstract
The overactivation of Janus kinases 2 (JAK2) by gain-of-function mutations in the JAK2, Myeloproliferative leukemia virus oncogene, or Calreticulin genes are the most important factor in the development of Philadelphia-negative myeloproliferative neoplasms (MPNs). The discovery of the JAK2V617F mutation is a significant breakthrough in understanding the pathogenesis of MPNs, and inhibition of JAK2 abnormal activation has become one of the most effective strategies against MPNs. Currently, three JAK2 inhibitors for treating MPNs have been approved, and several are being evaluated in clinical trials. However, persistent challenges in terms of drug resistance and off-target effects remain unresolved. In this review, we introduce and classify the available JAK2 inhibitors in terms of their mechanisms and clinical considerations. Additionally, through an analysis of target points, binding modes, and structure-activity inhibitor relationships, we propose strategies such as combination therapy and allosteric inhibitors to overcome specific challenges. This review offers valuable insights into current trends and future directions for optimal management of MPNs using JAK2 inhibitors.
Collapse
Affiliation(s)
- Xiaofeng Liu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Binyou Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
- Zigong Mental Health Center, Zigong Affiliated Hospital of Southwest Medical University, Zigong, 643000, China
| | - Yuan Liu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Yang Yu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
- Zigong Mental Health Center, Zigong Affiliated Hospital of Southwest Medical University, Zigong, 643000, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Jianming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- Zigong Mental Health Center, Zigong Affiliated Hospital of Southwest Medical University, Zigong, 643000, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Yiwei Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- Zigong Mental Health Center, Zigong Affiliated Hospital of Southwest Medical University, Zigong, 643000, China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
3
|
Berenson JR, Limon A, Rice S, Safaie T, Boccia R, Yang H, Moezi M, Lim S, Schwartz G, Eshaghian S, Brobeck M, Swift R, Eades BM, Bujarski S, Sebhat Y, Ray R, Kim S, Del Dosso A, Vescio R. A Phase I Trial Evaluating the Addition of Lenalidomide to Patients with Relapsed/Refractory Multiple Myeloma Progressing on Ruxolitinib and Methylprednisolone. Target Oncol 2024; 19:343-357. [PMID: 38643346 DOI: 10.1007/s11523-024-01049-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Ruxolitinib (RUX), an orally administered selective Janus kinase 1/2 inhibitor, has received approval for the treatment of myelofibrosis, polycythemia vera, and graft-versus-host disease. We have previously demonstrated the anti-multiple myeloma effects of RUX alone and in combination with the immunomodulatory agent lenalidomide (LEN) and glucocorticosteroids both pre-clinically and clinically. OBJECTIVE This study aims to evaluate whether LEN can achieve clinical activity among patients with multiple myeloma progressing on the combination of RUX and methylprednisolone (MP). METHODS In this part of a phase I, multicenter, open-label study, we evaluated the safety and efficacy of RUX and MP for patients with multiple myeloma with progressive disease who had previously received a proteasome inhibitor, LEN, glucocorticosteroids, and at least three prior regimens; we also determined the safety and efficacy of adding LEN at the time of disease progression from the initial doublet treatment. Initially, all subjects received oral RUX 15 mg twice daily and oral MP 40 mg every other day. Those patients who developed progressive disease according to the International Myeloma Working Group criteria then received LEN 10 mg once daily on days 1-21 within a 28-day cycle in addition to RUX and MP, which were administered at the same doses these patients were receiving at the time progressive disease developed. RESULTS Twenty-nine subjects (median age 64 years; 18 [62%] male) were enrolled in this part of the study and initially received the two-drug combination of RUX and MP. The median number of prior therapies was six (range 3-12). The overall response rate from this two-drug combination was 31% and the clinical benefit rate was 34%. The best responses were 1 very good partial response, 8 partial responses, 1 minor response, 12 stable disease, and 7 progressive disease. The median progression-free survival was 3.5 months (range 0.5-36.2 months). The median time to response was 3.0 months. The median duration of response was 12.5 months (range 2.8-36.2 months). Twenty (69%) patients who showed progressive disease had LEN added to RUX and MP; all patients had prior exposure to LEN and all but one patient was refractory to their last LEN-containing regimen. After the addition of LEN, the overall response rate was 30% and the clinical benefit rate was 40%. The best responses of patients following the addition of LEN were 2 very good partial responses, 4 partial responses, 2 minor responses, 8 stable disease, and 4 progressive disease. The median time to response was 2.6 months (range 0.7-15.0 months). The median duration of response was not reached. The median progression-free survival following the addition of LEN was 3.5 months (range 0.3-25.9 months). CONCLUSIONS For patients with multiple myeloma, treatment with RUX and MP is effective and well tolerated, and LEN can be used to extend the benefit of this RUX-based treatment. CLINICAL TRIAL REGISTRATION This study is registered with ClinicalTrials.gov, NCT03110822, and is ongoing.
Collapse
Affiliation(s)
- James R Berenson
- Institute for Myeloma & Bone Cancer Research, West Hollywood, CA, USA.
- ONCOtherapeutics, 9201 Sunset Boulevard Suite 300, West Hollywood, CA, 90069, USA.
- Berenson Cancer Center, West Hollywood, CA, USA.
| | - Andrea Limon
- ONCOtherapeutics, 9201 Sunset Boulevard Suite 300, West Hollywood, CA, 90069, USA
| | - Stephanie Rice
- ONCOtherapeutics, 9201 Sunset Boulevard Suite 300, West Hollywood, CA, 90069, USA
| | - Tahmineh Safaie
- ONCOtherapeutics, 9201 Sunset Boulevard Suite 300, West Hollywood, CA, 90069, USA
| | - Ralph Boccia
- Center for Cancer and Blood Disorders, Bethesda, MD, USA
| | - Honghao Yang
- The Oncology Institute of Hope and Innovation, Alhambra, CA, USA
| | - Mehdi Moezi
- Cancer Specialists of North Florida, Fleming Island, FL, USA
| | - Stephen Lim
- Cedars Sinai Samuel Oschin Cancer Center, Los Angeles, CA, USA
| | | | | | - Matthew Brobeck
- ONCOtherapeutics, 9201 Sunset Boulevard Suite 300, West Hollywood, CA, 90069, USA
| | | | | | | | | | - Rudra Ray
- Berenson Cancer Center, West Hollywood, CA, USA
| | - Susanna Kim
- ONCOtherapeutics, 9201 Sunset Boulevard Suite 300, West Hollywood, CA, 90069, USA
| | - Ashley Del Dosso
- ONCOtherapeutics, 9201 Sunset Boulevard Suite 300, West Hollywood, CA, 90069, USA
| | - Robert Vescio
- Cedars Sinai Samuel Oschin Cancer Center, Los Angeles, CA, USA
| |
Collapse
|
4
|
Del Dosso A, Tadevosyan E, Berenson JR. Preclinical and clinical evaluation of the Janus Kinase inhibitor ruxolitinib in multiple myeloma. Oncotarget 2024; 15:65-75. [PMID: 38319731 PMCID: PMC10852065 DOI: 10.18632/oncotarget.28547] [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: 11/27/2023] [Accepted: 12/18/2023] [Indexed: 02/08/2024] Open
Abstract
Multiple myeloma (MM) is the most common primary malignancy of the bone marrow. No established curative treatment is currently available for patients diagnosed with MM. In recent years, new and more effective drugs have become available for the treatment of this B-cell malignancy. These new drugs have often been evaluated together and in combination with older agents. However, even these novel combinations eventually become ineffective; and, thus, novel therapeutic approaches are necessary to help overcome resistance to these treatments. Recently, the Janus Kinase (JAK) family of tyrosine kinases, specifically JAK1 and JAK2, has been shown to have a role in the pathogenesis of MM. Preclinical studies have demonstrated a role for JAK signaling in direct and indirect growth of MM and downregulation of anti-tumor immune responses in these patients. Also, inhibition of JAK proteins enhances the anti-MM effects of other drugs used to treat MM. These findings have been confirmed in clinical studies which have further demonstrated the safety and efficacy of JAK inhibition as a means to overcome resistance to currently available anti-MM therapies. Additional studies will provide further support for this promising new therapeutic approach for treating patients with MM.
Collapse
Affiliation(s)
- Ashley Del Dosso
- ONCOtherapeutics, West Hollywood, CA 90069, USA
- These authors contributed equally to this work
| | - Elizabeth Tadevosyan
- Berenson Cancer Center, West Hollywood, CA 90069, USA
- These authors contributed equally to this work
| | - James R. Berenson
- ONCOtherapeutics, West Hollywood, CA 90069, USA
- Berenson Cancer Center, West Hollywood, CA 90069, USA
- Institute for Myeloma and Bone Cancer Research, West Hollywood, CA 90069, USA
| |
Collapse
|
5
|
Liu Y, Wang W, Zhang J, Gao S, Xu T, Yin Y. JAK/STAT signaling in diabetic kidney disease. Front Cell Dev Biol 2023; 11:1233259. [PMID: 37635867 PMCID: PMC10450957 DOI: 10.3389/fcell.2023.1233259] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Diabetic kidney disease (DKD) is the most important microvascular complication of diabetes and the leading cause of end-stage renal disease (ESRD) worldwide. The Janus kinase/signal transducer and activator of the transcription (JAK/STAT) signaling pathway, which is out of balance in the context of DKD, acts through a range of metabolism-related cytokines and hormones. JAK/STAT is the primary signaling node in the progression of DKD. The latest research on JAK/STAT signaling helps determine the role of this pathway in the factors associated with DKD progression. These factors include the renin-angiotensin system (RAS), fibrosis, immunity, inflammation, aging, autophagy, and EMT. This review epitomizes the progress in understanding the complicated explanation of the etiologies of DKD and the role of the JAK/STAT pathway in the progression of DKD and discusses whether it can be a potential target for treating DKD. It further summarizes the JAK/STAT inhibitors, natural products, and other drugs that are promising for treating DKD and discusses how these inhibitors can alleviate DKD to explore possible potential drugs that will contribute to formulating effective treatment strategies for DKD in the near future.
Collapse
Affiliation(s)
- Yingjun Liu
- Clinical Medicine Department, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenkuan Wang
- Clinical Medicine Department, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jintao Zhang
- Clinical Medicine Department, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuo Gao
- Clinical Medicine Department, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tingting Xu
- Clinical Medicine Department, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yonghui Yin
- Department of Endocrinology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
6
|
Tian Y, Qin S, Zhang F, Luo J, He X, Sun Y, Yang T. Discovery of N-(4-(Aminomethyl)phenyl)-5-methylpyrimidin-2-amine Derivatives as Potent and Selective JAK2 Inhibitors. ACS Med Chem Lett 2023; 14:1113-1121. [PMID: 37583815 PMCID: PMC10424325 DOI: 10.1021/acsmedchemlett.3c00251] [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: 06/05/2023] [Accepted: 07/28/2023] [Indexed: 08/17/2023] Open
Abstract
The JAK2V617F mutation leads to JAK2 autophosphorylation and activation of downstream pathways, eventually resulting in myeloproliferative neoplasms (MPNs). Selective inhibitors showed advantages in terms of side effects; therefore, there is an urgent need to develop novel selective JAK2 inhibitors for treating MPNs. In this study, we described a series of N-(4-(aminomethyl)phenyl)pyrimidin-2-amine derivatives as selective JAK2 inhibitors. Systematic exploration through opening the tetrahydroisoquinoline based on the previous lead compound 13ac led to the discovery of the optimal compound A8. Compound A8 showed excellent potency on JAK2 kinase, with an IC50 value of 5 nM, and inhibited the phosphorylation of JAK2 and its downstream signaling pathway. Moreover, A8 exhibited 38.6-, 54.6-, and 41.2-fold selectivity for JAK1, JAK3, and TYK2, respectively. Compared to the lead compound, A8 demonstrated much better metabolic stabilities, with a bioavailability of 41.1%. These findings suggest that A8 is a relatively selective JAK2 inhibitor, deserving to be developed for treating MPNs.
Collapse
Affiliation(s)
- Yang Tian
- Department
of Otolaryngology Head and Neck Surgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital
of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated
to Chongqing Medical University, Chengdu 610014, China
- Medical
Research Center. The Third People’s Hospital of Chengdu, The
Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu
Hospital Affiliated to Chongqing Medical University, Chengdu, 610014, China
| | - Songhui Qin
- State
Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation
Center of Biotherapy, Chengdu 610041, China
| | - Fang Zhang
- Department
of Otolaryngology Head and Neck Surgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital
of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated
to Chongqing Medical University, Chengdu 610014, China
| | - Jing Luo
- Department
of Otolaryngology Head and Neck Surgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital
of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated
to Chongqing Medical University, Chengdu 610014, China
| | - Xi He
- Department
of Otolaryngology Head and Neck Surgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital
of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated
to Chongqing Medical University, Chengdu 610014, China
| | - Yi Sun
- Department
of Otolaryngology Head and Neck Surgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital
of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated
to Chongqing Medical University, Chengdu 610014, China
| | - Tao Yang
- State
Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation
Center of Biotherapy, Chengdu 610041, China
| |
Collapse
|
7
|
Algeri M, Becilli M, Locatelli F. Ruxolitinib as the first post-steroid treatment for acute and chronic graft-versus-host disease. Expert Rev Clin Immunol 2023; 19:1299-1313. [PMID: 37606511 DOI: 10.1080/1744666x.2023.2249230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 08/14/2023] [Indexed: 08/23/2023]
Abstract
INTRODUCTION Acute and chronic graft-versus-host disease (GvHD) are potentially life-threatening complications occurring after allogeneic stem cell transplantation (allo-HSCT). Although steroids represent the first-line treatment for both conditions, in those patients who do not adequately benefit from steroid therapy, standardized treatment algorithms are lacking. In recent years, ruxolitinib has emerged as the most promising agent for the second-line therapy of steroid-refractory (SR)-GvHD. AREAS COVERED This review will summarize the biological properties and the mechanistic aspects that justify the therapeutic role of ruxolitinib in GvHD. In addition, current treatment options for SR-GvHD will be briefly discussed. Finally, results of the most relevant clinical trials on the use of ruxolitinib for SR-GvHD will be analyzed, with a particular focus on two phase-III randomized trials in which ruxolitinib demonstrated its superiority in comparison with the best available therapy. EXPERT OPINION Ruxolitinib has considerably improved the outcome of patients with SR-acute/chronic-GvHD and should be regarded as the standard-of-care option when corticosteroids fail or cannot be tapered. Nevertheless, a number of questions still remain unanswered and significant room for improvement exists. Additional observations derived from a longer follow-up will certainly increase our expertise in the management of this powerful therapy.
Collapse
Affiliation(s)
- Mattia Algeri
- Department of Haematology/Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Health Science, Magna Grecia University of Catanzaro, Catanzaro, Italy
| | - Marco Becilli
- Department of Haematology/Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Franco Locatelli
- Department of Haematology/Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
8
|
Gaido OER, Pavlaki N, Granger JM, Mesubi OO, Liu B, Lin BL, Long A, Walker D, Mayourian J, Schole KL, Terrillion CE, Nkashama LJ, Hulsurkar MM, Dorn LE, Ferrero KM, Huganir RL, Müller FU, Wehrens XHT, Liu JO, Luczak ED, Bezzerides VJ, Anderson ME. An improved reporter identifies ruxolitinib as a potent and cardioprotective CaMKII inhibitor. Sci Transl Med 2023; 15:eabq7839. [PMID: 37343080 PMCID: PMC11022683 DOI: 10.1126/scitranslmed.abq7839] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 05/31/2023] [Indexed: 06/23/2023]
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) hyperactivity causes cardiac arrhythmias, a major source of morbidity and mortality worldwide. Despite proven benefits of CaMKII inhibition in numerous preclinical models of heart disease, translation of CaMKII antagonists into humans has been stymied by low potency, toxicity, and an enduring concern for adverse effects on cognition due to an established role of CaMKII in learning and memory. To address these challenges, we asked whether any clinically approved drugs, developed for other purposes, were potent CaMKII inhibitors. For this, we engineered an improved fluorescent reporter, CaMKAR (CaMKII activity reporter), which features superior sensitivity, kinetics, and tractability for high-throughput screening. Using this tool, we carried out a drug repurposing screen (4475 compounds in clinical use) in human cells expressing constitutively active CaMKII. This yielded five previously unrecognized CaMKII inhibitors with clinically relevant potency: ruxolitinib, baricitinib, silmitasertib, crenolanib, and abemaciclib. We found that ruxolitinib, an orally bioavailable and U.S. Food and Drug Administration-approved medication, inhibited CaMKII in cultured cardiomyocytes and in mice. Ruxolitinib abolished arrhythmogenesis in mouse and patient-derived models of CaMKII-driven arrhythmias. A 10-min pretreatment in vivo was sufficient to prevent catecholaminergic polymorphic ventricular tachycardia, a congenital source of pediatric cardiac arrest, and rescue atrial fibrillation, the most common clinical arrhythmia. At cardioprotective doses, ruxolitinib-treated mice did not show any adverse effects in established cognitive assays. Our results support further clinical investigation of ruxolitinib as a potential treatment for cardiac indications.
Collapse
Affiliation(s)
- Oscar E. Reyes Gaido
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nikoleta Pavlaki
- Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan M. Granger
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Olurotimi O. Mesubi
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bian Liu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Brian L. Lin
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alan Long
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David Walker
- Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Joshua Mayourian
- Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kate L. Schole
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chantelle E. Terrillion
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lubika J. Nkashama
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mohit M. Hulsurkar
- Cardiovascular Research Institute and Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lauren E. Dorn
- Cardiovascular Research Institute and Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kimberly M. Ferrero
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L. Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Frank U. Müller
- Institute of Pharmacology and Toxicology, University of Münster, Münster 48149, Germany
| | - Xander H. T. Wehrens
- Cardiovascular Research Institute and Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Departments of Medicine, Neuroscience, and Pediatrics, Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jun O. Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Elizabeth D. Luczak
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Vassilios J. Bezzerides
- Department of Cardiology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mark E. Anderson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Division of Biological Sciences and the Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
9
|
Appeldoorn TYJ, Munnink THO, Morsink LM, Hooge MNLD, Touw DJ. Pharmacokinetics and Pharmacodynamics of Ruxolitinib: A Review. Clin Pharmacokinet 2023; 62:559-571. [PMID: 37000342 PMCID: PMC10064968 DOI: 10.1007/s40262-023-01225-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND AND OBJECTIVE Ruxolitinib is a tyrosine kinase inhibitor targeting the Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathways. Ruxolitinib is used to treat myelofibrosis, polycythemia vera and steroid-refractory graft-versus-host disease in the setting of allogeneic stem-cell transplantation. This review describes the pharmacokinetics and pharmacodynamics of ruxolitinib. METHODS Pubmed, EMBASE, Cochrane Library and web of Science were searched from the time of database inception to march 15, 2021 and was repeated on November 16, 2021. Articles not written in English, animal or in vitro studies, letters to the editor, case reports, where ruxolitinib was not used for hematological diseases or not available as full text were excluded. RESULTS Ruxolitinib is well absorbed, has 95% bio-availability, and is bound to albumin for 97%. Ruxolitinib pharmacokinetics can be described with a two-compartment model and linear elimination. Volume of distribution differs between men and women, likely related to bodyweight differences. Metabolism is mainly hepatic via CYP3A4 and can be altered by CYP3A4 inducers and inhibitors. The major metabolites of ruxolitinib are pharmacologically active. The main route of elimination of ruxolitinib metabolites is renal. Liver and renal dysfunction affect some of the pharmacokinetic variables and require dose reductions. Model-informed precision dosing might be a way to further optimize and individualize ruxolitinib treatment, but is not yet advised for routine care due to lack of information on target concentrations. CONCLUSION Further research is needed to explain the interindividual variability of the ruxolitinib pharmacokinetic variables and to optimize individual treatment.
Collapse
Affiliation(s)
- T Y J Appeldoorn
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - T H Oude Munnink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - L M Morsink
- Department of Hematology, University Medical Centre Groningen, Groningen, The Netherlands
| | - M N Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands
| | - D J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
10
|
Borgström EW, Edvinsson M, Pérez LP, Norlin AC, Enoksson SL, Hansen S, Fasth A, Friman V, Kämpe O, Månsson R, Estupiñán HY, Wang Q, Ziyang T, Lakshmikanth T, Smith CIE, Brodin P, Bergman P. Three Adult Cases of STAT1 Gain-of-Function with Chronic Mucocutaneous Candidiasis Treated with JAK Inhibitors. J Clin Immunol 2023; 43:136-150. [PMID: 36050429 PMCID: PMC9840596 DOI: 10.1007/s10875-022-01351-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/08/2022] [Indexed: 01/21/2023]
Abstract
PURPOSE The aim of this study was to characterize clinical effects and biomarkers in three patients with chronic mucocutaneous candidiasis (CMC) caused by gain-of-function (GOF) mutations in the STAT1 gene during treatment with Janus kinase (JAK) inhibitors. METHODS Mass cytometry (CyTOF) was used to characterize mononuclear leukocyte populations and Olink assay to quantify 265 plasma proteins. Flow-cytometric Assay for Specific Cell-mediated Immune-response in Activated whole blood (FASCIA) was used to quantify the reactivity against Candida albicans. RESULTS Overall, JAK inhibitors improved clinical symptoms of CMC, but caused side effects in two patients. Absolute numbers of neutrophils, T cells, B cells, and NK cells were sustained during baricitinib treatment. Detailed analysis of cellular subsets, using CyTOF, revealed increased expression of CD45, CD52, and CD99 in NK cells, reflecting a more functional phenotype. Conversely, monocytes and eosinophils downregulated CD16, consistent with reduced inflammation. Moreover, T and B cells showed increased expression of activation markers during treatment. In one patient with a remarkable clinical effect of baricitinib treatment, the immune response to C. albicans increased after 7 weeks of treatment. Alterations in plasma biomarkers involved downregulation of cellular markers CXCL10, annexin A1, granzyme B, granzyme H, and oncostatin M, whereas FGF21 was the only upregulated marker after 7 weeks. After 3 months, IFN-ɣ and CXCL10 were downregulated. CONCLUSIONS The clinical effect of JAK inhibitor treatment of CMC is promising. Several biological variables were altered during baricitinib treatment demonstrating that lymphocytes, NK cells, monocytes, and eosinophils were affected. In parallel, cellular reactivity against C. albicans was enhanced.
Collapse
Affiliation(s)
- Emilie W. Borgström
- Department of Laboratory Medicine, Clinical Microbiology, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Marie Edvinsson
- grid.412354.50000 0001 2351 3333Department of Medical Sciences, Section of Infectious Diseases, Uppsala University Hospital, Uppsala, Sweden
| | - Lucía P. Pérez
- grid.4714.60000 0004 1937 0626Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna C. Norlin
- grid.24381.3c0000 0000 9241 5705Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sara L. Enoksson
- grid.24381.3c0000 0000 9241 5705Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Susanne Hansen
- grid.24381.3c0000 0000 9241 5705Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Fasth
- grid.8761.80000 0000 9919 9582Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Vanda Friman
- grid.8761.80000 0000 9919 9582Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olle Kämpe
- grid.4714.60000 0004 1937 0626Experimental Endocrinology, Department of Medicine, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Robert Månsson
- grid.4714.60000 0004 1937 0626Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hernando Y. Estupiñán
- grid.4714.60000 0004 1937 0626Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden ,grid.411595.d0000 0001 2105 7207Departamento de Ciencias Básicas, Universidad Industrial de Santander, 680002 Bucaramanga, Colombia
| | - Qing Wang
- grid.4714.60000 0004 1937 0626Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tan Ziyang
- grid.4714.60000 0004 1937 0626Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Tadepally Lakshmikanth
- grid.4714.60000 0004 1937 0626Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Carl Inge E. Smith
- grid.24381.3c0000 0000 9241 5705Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden ,Department of Laboratory Medicine, Translational Research Center Karolinska (TRACK), Stockholm, Sweden
| | - Petter Brodin
- grid.4714.60000 0004 1937 0626Science for Life Laboratory, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden ,grid.7445.20000 0001 2113 8111Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Peter Bergman
- Department of Laboratory Medicine, Clinical Microbiology, Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
11
|
Agashe RP, Lippman SM, Kurzrock R. JAK: Not Just Another Kinase. Mol Cancer Ther 2022; 21:1757-1764. [PMID: 36252553 PMCID: PMC10441554 DOI: 10.1158/1535-7163.mct-22-0323] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/07/2022] [Accepted: 10/07/2022] [Indexed: 01/12/2023]
Abstract
The JAK/STAT axis is implicated in cancer, inflammation, and immunity. Numerous cytokines/growth factors affect JAK/STAT signaling. JAKs (JAK1, JAK2, JAK3, and TYK2) noncovalently associate with cytokine receptors, mediate receptor tyrosine phosphorylation, and recruit ≥1 STAT proteins (STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6). Tyrosine-phosphorylated STATs dimerize and are then transported into the nucleus to function as transcription factors. Signaling is attenuated by specific suppressor of cytokine signaling proteins, creating a negative feedback loop. Both germline mutations and polymorphisms of JAK family members correlate with specific diseases: Systemic lupus erythematosus (TYK2 polymorphisms); severe combined immunodeficiency (JAK3 mutations); pediatric acute lymphoblastic leukemia (TYK2 mutations); and hereditary thrombocytosis (JAK2 mutations). Somatic gain-of-function JAK mutations mainly occur in hematologic malignancies, with the activating JAK2 V617F being a myeloproliferative disorder hallmark; it is also seen in clonal hematopoiesis of indeterminate potential. Several T-cell malignancies, as well as B-cell acute lymphoblastic leukemia, and acute megakaryoblastic leukemia also harbor JAK family somatic alterations. On the other hand, JAK2 copy-number loss is associated with immune checkpoint inhibitor resistance. JAK inhibitors (jakinibs) have been deployed in many conditions with JAK activation; they are approved in myeloproliferative disorders, rheumatoid and psoriatic arthritis, atopic dermatitis, ulcerative colitis, graft-versus-host disease, alopecia areata, ankylosing spondylitis, and in patients hospitalized for COVID-19. Clinical trials are investigating jakinibs in multiple other autoimmune/inflammatory conditions. Furthermore, dermatologic and neurologic improvements have been observed in children with Aicardi-Goutieres syndrome (a genetic interferonopathy) treated with JAK inhibitors.
Collapse
Affiliation(s)
| | | | - Razelle Kurzrock
- Medical College of Wisconsin, Milwaukee, Wisconsin
- Win Consortium, Paris, France
| |
Collapse
|
12
|
Zheng TJ, Parra-Izquierdo I, Reitsma SE, Heinrich MC, Larson MK, Shatzel JJ, Aslan JE, McCarty OJT. Platelets and tyrosine kinase inhibitors: clinical features, mechanisms of action, and effects on physiology. Am J Physiol Cell Physiol 2022; 323:C1231-C1250. [PMID: 35938677 PMCID: PMC9576167 DOI: 10.1152/ajpcell.00040.2022] [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: 02/01/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) have emerged as a promising class of target-directed, small molecule inhibitors used to treat hematologic malignancies, inflammatory diseases, and autoimmune disorders. Recently, TKIs have also gained interest as potential antiplatelet-directed therapeutics that could be leveraged to reduce pathologic thrombus formation and atherothrombotic complications, while minimally affecting platelet hemostatic function. This review provides a mechanistic overview and summarizes the known effects of tyrosine kinase inhibitors on platelet signaling and function, detailing prominent platelet signaling pathways downstream of the glycoprotein VI (GPVI) receptor, integrin αIIbβ3, and G protein-coupled receptors (GPCRs). This review focuses on mechanistic as well as clinically relevant and emerging TKIs targeting major families of tyrosine kinases including but not limited to Bruton's tyrosine kinase (BTK), spleen tyrosine kinase (Syk), Src family kinases (SFKs), Janus kinases (JAK), and signal transducers and activators of transcription (STAT) and evaluates their effects on platelet aggregation and adhesion, granule secretion, receptor expression and activation, and protein phosphorylation events. In summation, this review highlights current advances and knowledge on the effects of select TKIs on platelet biology and furthers insight on signaling pathways that may represent novel druggable targets coupled to specific platelet functional responses.
Collapse
Affiliation(s)
- Tony J Zheng
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Iván Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Stéphanie E Reitsma
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Michael C Heinrich
- Portland Veterans Affairs Health Care System and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Molecular and Cellular Biosciences, Oregon Health & Science University, Portland, Oregon
| | - Mark K Larson
- Department of Biology, Augustana University, Sioux Falls, South Dakota
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental & Cancer Biology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
13
|
Di Raimondo C, Rao L, Lozzi F, Lombardo P, Silvaggio D, Vellucci L, Tofani L, Campione E, Bianchi L. Cemiplimab and ruxolitinib in concomitant cutaneous squamous cell carcinoma and myelofibrosis. Dermatol Ther 2022; 35:e15421. [DOI: 10.1111/dth.15421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/02/2022] [Indexed: 11/29/2022]
Affiliation(s)
| | - Ludovico Rao
- Department of Dermatology University of Roma Tor Vergata Rome Italy
| | - Flavia Lozzi
- Department of Dermatology University of Roma Tor Vergata Rome Italy
| | - Paolo Lombardo
- Department of Dermatology University of Roma Tor Vergata Rome Italy
| | | | - Laura Vellucci
- Department of Dermatology University of Roma Tor Vergata Rome Italy
| | - Lorenzo Tofani
- Department of Dermatology University of Roma Tor Vergata Rome Italy
| | - Elena Campione
- Department of Dermatology University of Roma Tor Vergata Rome Italy
| | - Luca Bianchi
- Department of Dermatology University of Roma Tor Vergata Rome Italy
| |
Collapse
|
14
|
Simmons DP, Nguyen HN, Gomez-Rivas E, Jeong Y, Jonsson AH, Chen AF, Lange JK, Dyer GS, Blazar P, Earp BE, Coblyn JS, Massarotti EM, Sparks JA, Todd DJ, Accelerating Medicines Partnership ® (AMP ®) RA/SLE Network, Rao DA, Kim EY, Brenner MB. SLAMF7 engagement superactivates macrophages in acute and chronic inflammation. Sci Immunol 2022; 7:eabf2846. [PMID: 35148199 PMCID: PMC8991457 DOI: 10.1126/sciimmunol.abf2846] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophages regulate protective immune responses to infectious microbes, but aberrant macrophage activation frequently drives pathological inflammation. To identify regulators of vigorous macrophage activation, we analyzed RNA-seq data from synovial macrophages and identified SLAMF7 as a receptor associated with a superactivated macrophage state in rheumatoid arthritis. We implicated IFN-γ as a key regulator of SLAMF7 expression and engaging SLAMF7 drove a strong wave of inflammatory cytokine expression. Induction of TNF-α after SLAMF7 engagement amplified inflammation through an autocrine signaling loop. We observed SLAMF7-induced gene programs not only in macrophages from rheumatoid arthritis patients but also in gut macrophages from patients with active Crohn's disease and in lung macrophages from patients with severe COVID-19. This suggests a central role for SLAMF7 in macrophage superactivation with broad implications in human disease pathology.
Collapse
Affiliation(s)
- Daimon P. Simmons
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Hung N. Nguyen
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Emma Gomez-Rivas
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Yunju Jeong
- Harvard Medical School, Boston, MA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - A. Helena Jonsson
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Antonia F. Chen
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Jeffrey K. Lange
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA
| | - George S. Dyer
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Philip Blazar
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Brandon E. Earp
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Boston, MA
| | - Jonathan S. Coblyn
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Elena M. Massarotti
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Jeffrey A. Sparks
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Derrick J. Todd
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | | | - Deepak A. Rao
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Edy Y. Kim
- Harvard Medical School, Boston, MA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Michael B. Brenner
- Harvard Medical School, Boston, MA
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
15
|
Yang T, Cui X, Tang M, Qi W, Zhu Z, Shi M, Yang L, Pei H, Zhang W, Xie L, Xu Y, Yang Z, Chen L. Identification of a Novel 2,8-Diazaspiro[4.5]decan-1-one Derivative as a Potent and Selective Dual TYK2/JAK1 Inhibitor for the Treatment of Inflammatory Bowel Disease. J Med Chem 2022; 65:3151-3172. [PMID: 35113547 DOI: 10.1021/acs.jmedchem.1c01137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this study, we described a series of 2,8-diazaspiro[4.5]decan-1-one derivatives as selective TYK2/JAK1 inhibitors. Systematic exploration of the structure-activity relationship through the introduction of spirocyclic scaffolds based on the reported selective TYK2 inhibitor 14l led to the discovery of the superior derivative compound 48. Compound 48 showed excellent potency on TYK2/JAK1 kinases with IC50 values of 6 and 37 nM, respectively, and exhibited more than 23-fold selectivity for JAK2. Compound 48 also demonstrated excellent metabolic stability and more potent anti-inflammatory efficacy than tofacitinib in acute ulcerative colitis models. Moreover, the excellent anti-inflammatory effect of compound 48 was mediated by regulating the expression of related TYK2/JAK1-regulated genes, as well as the formation of Th1, Th2, and Th17 cells. Taken together, these findings suggest that compound 48 is a selective dual TYK2/JAK inhibitor, deserving to be developed as a clinical candidate.
Collapse
Affiliation(s)
- Tao Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Xue Cui
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Wenyan Qi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Zejiang Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Mingsong Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Linyu Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Heying Pei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Wanhua Zhang
- Department of Hematology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Lixin Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yaohui Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China.,Chengdu Zenitar Biomedical Technology Co., Ltd., Chengdu 610041, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China.,Chengdu Zenitar Biomedical Technology Co., Ltd., Chengdu 610041, China
| |
Collapse
|
16
|
Chifotides HT, Bose P, Verstovsek S. Momelotinib: an emerging treatment for myelofibrosis patients with anemia. J Hematol Oncol 2022; 15:7. [PMID: 35045875 PMCID: PMC8772195 DOI: 10.1186/s13045-021-01157-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/30/2021] [Indexed: 12/11/2022] Open
Abstract
The suite of marked anemia benefits that momelotinib has consistently conferred on myelofibrosis (MF) patients stem from its unique inhibitory activity on the BMP6/ACVR1/SMAD and IL-6/JAK/STAT3 pathways, resulting in decreased hepcidin (master iron regulator) expression, higher serum iron and hemoglobin levels, and restored erythropoiesis. Clinical data on momelotinib from the phase 2 and the two phase 3 SIMPLIFY trials consistently demonstrated high rates of sustained transfusion-independence. In a recent phase 2 translational study, 41% of the patients achieved transfusion independence for ≥ 12 weeks. In the phase 3 trials SIMPLIFY-1 and SIMPLIFY-2, 17% more JAK inhibitor-naïve patients and two-fold more JAK inhibitor-treated patients achieved or maintained transfusion independence with momelotinib versus ruxolitinib and best available therapy (89% ruxolitinib), respectively. Anemia is present in approximately a third of MF patients at diagnosis, eventually developing in nearly all patients. The need for red blood cell transfusions is an independent adverse risk factor for both overall survival and leukemic transformation. Presently, FDA-approved medications to address anemia are lacking. Momelotinib is one of the prime candidates to durably address the critical unmet needs of MF patients with moderate/severe anemia. Importantly, momelotinib may have overall survival benefits in frontline and second-line MF patients. MOMENTUM is an international registration-track phase 3 trial further assessing momelotinib’s unique constellation of anemia and other benefits in second-line MF patients; the results of the MOMENTUM trial are keenly awaited and may lead to regulatory approval of momelotinib.
Collapse
|
17
|
Suppa M, Marino L, Piccari P, Masselli G, Gradini R. Cardiac complications in a patient affected by systemic mastocytosis and primitive myelofibrosis: A case report. Clin Case Rep 2021; 9:e04972. [PMID: 34804526 PMCID: PMC8587177 DOI: 10.1002/ccr3.4972] [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: 07/14/2021] [Accepted: 10/04/2021] [Indexed: 11/18/2022] Open
Abstract
Systemic mastocytosis with associated primitive myelofibrosis is a rare and complex disease with a difficult therapeutic management. The release of several inflammation mediators can trigger acute cardiovascular events.
Collapse
Affiliation(s)
- Marianna Suppa
- Emergency DepartmentUmberto I HospitalUniversity Sapienza of RomeRomeItaly
| | - Luca Marino
- Emergency DepartmentUmberto I HospitalUniversity Sapienza of RomeRomeItaly
- Mechanical and Aerospace DepartmentUniversity Sapienza of RomeRomeItaly
| | - Pietro Piccari
- Emergency DepartmentUmberto I HospitalUniversity Sapienza of RomeRomeItaly
| | - Gabriele Masselli
- Radiology DepartmentUmberto I HospitalUniversity Sapienza of RomeRomeItaly
| | - Roberto Gradini
- Department of Experimental MedicineUniversity Sapienza of RomeRomeItaly
| |
Collapse
|
18
|
Parra-Izquierdo I, Melrose AR, Pang J, Lakshmanan HHS, Reitsma SE, Vavilapalli SH, Larson MK, Shatzel JJ, McCarty OJT, Aslan JE. Janus kinase inhibitors ruxolitinib and baricitinib impair glycoprotein-VI mediated platelet function. Platelets 2021; 33:404-415. [PMID: 34097573 PMCID: PMC8648864 DOI: 10.1080/09537104.2021.1934665] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Several Janus kinase (JAK) inhibitors (jakinibs) have recently been approved to treat inflammatory, autoimmune and hematological conditions. Despite emerging roles for JAKs and downstream signal transducer and activator of transcription (STAT) proteins in platelets, it remains unknown whether jakinibs affect platelet function. Here, we profile platelet biochemical and physiological responses in vitro in the presence of five different clinically relevant jakinibs, including ruxolitinib, upadacitinib, oclacitinib, baricitinib and tofacitinib. Flow cytometry, microscopy and other assays found that potent JAK1/2 inhibitors baricitinib and ruxolitinib reduced platelet adhesion to collagen, as well as platelet aggregation, secretion and integrin αIIbβ3 activation in response to the glycoprotein VI (GPVI) agonist collagen-related peptide (CRP-XL). Western blot analysis demonstrated that jakinibs reduced Akt phosphorylation and activation following GPVI activation, where ruxolitinib and baricitinib prevented DAPP1 phosphorylation. In contrast, jakinibs had no effects on platelet responses to thrombin. Inhibitors of GPVI and JAK signaling also abrogated platelet STAT5 phosphorylation following CRP-XL stimulation. Additional pharmacologic experiments supported roles for STAT5 in platelet secretion, integrin activation and cytoskeletal responses. Together, our results demonstrate that ruxolitinib and baricitinib have inhibitory effects on platelet function in vitro and support roles for JAK/STAT5 pathways in GPVI/ITAM mediated platelet function.
Collapse
Affiliation(s)
- Iván Parra-Izquierdo
- Knight Cardiovascular Institute and Division of Cardiology, Oregon Health & Science University, Portland, OR, USA.,Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Alexander R Melrose
- Knight Cardiovascular Institute and Division of Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | | | - Stéphanie E Reitsma
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Sai Hitesh Vavilapalli
- Knight Cardiovascular Institute and Division of Cardiology, Oregon Health & Science University, Portland, OR, USA
| | - Mark K Larson
- Biology Department, Augustana University, Sioux Falls, SD, USA
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Division of Hematology and Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Division of Hematology and Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Joseph E Aslan
- Knight Cardiovascular Institute and Division of Cardiology, Oregon Health & Science University, Portland, OR, USA.,Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
19
|
Garcia-Melendo C, Cubiró X, Puig L. Janus Kinase Inhibitors in Dermatology: Part 1 — General Considerations and Applications in Vitiligo and Alopecia Areata. ACTAS DERMO-SIFILIOGRAFICAS 2021. [DOI: 10.1016/j.adengl.2021.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
20
|
Inhibidores de JAK: usos en dermatología. Parte 1: generalidades, aplicaciones en vitíligo y en alopecia areata. ACTAS DERMO-SIFILIOGRAFICAS 2021. [DOI: 10.1016/j.ad.2020.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
21
|
Coltro G, Vannucchi AM. The safety of JAK kinase inhibitors for the treatment of myelofibrosis. Expert Opin Drug Saf 2020; 20:139-154. [PMID: 33327810 DOI: 10.1080/14740338.2021.1865912] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION During the last decade, the development of small molecule inhibitors of Janus kinases (JAKi) contributed to revolutionize the therapeutic landscape of myelofibrosis (MF). JAKi proved to be effective in controlling disease-related symptoms and splenomegaly with remarkable inter-drug variability. However, in some cases the border between clinical efficacy of JAKi and dose-dependent toxicities is narrow leading to sub-optimal dose modifications and/or treatment discontinuation. AREAS COVERED In the current review, the authors aimed at providing a comprehensive review of the safety profile of JAKi that are currently approved or in advanced clinical development. Also, a short discussion of promising JAKi in early clinical evaluation and molecules 'lost' early in clinical development is provided. Finally, we discuss the possible strategies aimed at strengthening the safety of JAKi while improving the therapeutic efficacy. EXPERT OPINION Overall, JAKi display a satisfactory risk-benefit ratio, with main toxicities being gastrointestinal or related to the myelo/immunosuppressive effects, generally mild and easily manageable. However, JAKi may be associated with potentially life-threatening toxicities, such as neurological and infectious events. Thus, many efforts are needed in order to optimize JAKi-based therapeutic strategies without burdening patient safety. This could be attempted through drug combinations or the development of more selective molecules.
Collapse
Affiliation(s)
- Giacomo Coltro
- Department of Clinical and Experimental Medicine, University of Florence , Florence, Italy.,CRIMM, Center of Research and Innovation for Myeloproliferative Neoplasms, AOU Careggi , Florence, Italy
| | - Alessandro M Vannucchi
- Department of Clinical and Experimental Medicine, University of Florence , Florence, Italy.,CRIMM, Center of Research and Innovation for Myeloproliferative Neoplasms, AOU Careggi , Florence, Italy
| |
Collapse
|
22
|
Damerau A, Gaber T, Ohrndorf S, Hoff P. JAK/STAT Activation: A General Mechanism for Bone Development, Homeostasis, and Regeneration. Int J Mol Sci 2020; 21:E9004. [PMID: 33256266 PMCID: PMC7729940 DOI: 10.3390/ijms21239004] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
The Janus kinase (JAK) signal transducer and activator of transcription (STAT) signaling pathway serves as an important downstream mediator for a variety of cytokines, hormones, and growth factors. Emerging evidence suggests JAK/STAT signaling pathway plays an important role in bone development, metabolism, and healing. In this light, pro-inflammatory cytokines are now clearly implicated in these processes as they can perturb normal bone remodeling through their action on osteoclasts and osteoblasts at both intra- and extra-articular skeletal sites. Here, we summarize the role of JAK/STAT pathway on development, homeostasis, and regeneration based on skeletal phenotype of individual JAK and STAT gene knockout models and selective inhibition of components of the JAK/STAT signaling including influences of JAK inhibition in osteoclasts, osteoblasts, and osteocytes.
Collapse
Affiliation(s)
- Alexandra Damerau
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany; (A.D.); (S.O.); (P.H.)
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Timo Gaber
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany; (A.D.); (S.O.); (P.H.)
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Sarah Ohrndorf
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany; (A.D.); (S.O.); (P.H.)
| | - Paula Hoff
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany; (A.D.); (S.O.); (P.H.)
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
- Endokrinologikum Berlin am Gendarmenmarkt, 10117 Berlin, Germany
| |
Collapse
|
23
|
Kouznetsov VV. COVID-19 treatment: Much research and testing, but far, few magic bullets against SARS-CoV-2 coronavirus. Eur J Med Chem 2020; 203:112647. [PMID: 32693298 PMCID: PMC7362854 DOI: 10.1016/j.ejmech.2020.112647] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022]
Abstract
The new virus of the of β-Coronaviruses genus, SARS-CoV-2, is the causative agent of coronavirus disease-2019 (COVID-19) and is winning a proverbial chess match against all players simultaneous, including physicians, clinicians, pathologists, doctors, scientists, economists, athletes and politicians. The COVID-19 outbreak has seriously threatened public health, killing the most vulnerable persons and causing general panic. To stop this disease, effective remedies (i.e., drugs, vaccines, personal protection elements, etc.) are urgently required. Unfortunately, no registered specific therapies (including antiviral therapies, immune-modulating agents and vaccines) are currently available to treat coronavirus infections, highlighting an urgent need for therapeutics targeting SARS-CoV-2. In this work, fourteen existing small molecule drugs or/and experimental drugs selected by experts and examined from the point of view of bioavailability via the Lipinski-Veber rules and assessment of their physicochemical descriptors. The aim of this study is to discover selected pattern similarities and peculiar characteristics that could be useful for antiviral drug optimization, drug combination or new antiviral agent design.
Collapse
|
24
|
Kouznetsov VV. COVID-19 treatment: Much research and testing, but far, few magic bullets against SARS-CoV-2 coronavirus. Eur J Med Chem 2020. [PMID: 32693298 DOI: 10.1016/j.ejmech.2020.112647.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The new virus of the of β-Coronaviruses genus, SARS-CoV-2, is the causative agent of coronavirus disease-2019 (COVID-19) and is winning a proverbial chess match against all players simultaneous, including physicians, clinicians, pathologists, doctors, scientists, economists, athletes and politicians. The COVID-19 outbreak has seriously threatened public health, killing the most vulnerable persons and causing general panic. To stop this disease, effective remedies (i.e., drugs, vaccines, personal protection elements, etc.) are urgently required. Unfortunately, no registered specific therapies (including antiviral therapies, immune-modulating agents and vaccines) are currently available to treat coronavirus infections, highlighting an urgent need for therapeutics targeting SARS-CoV-2. In this work, fourteen existing small molecule drugs or/and experimental drugs selected by experts and examined from the point of view of bioavailability via the Lipinski-Veber rules and assessment of their physicochemical descriptors. The aim of this study is to discover selected pattern similarities and peculiar characteristics that could be useful for antiviral drug optimization, drug combination or new antiviral agent design.
Collapse
Affiliation(s)
- Vladimir V Kouznetsov
- Laboratorio de Química Orgánica y Biomolecular, CMN, Universidad Industrial de Santander, Parque Tecnológico Guatiguará, Piedecuesta, 681011, Colombia.
| |
Collapse
|
25
|
Di Prima A, Botticelli A, Scalzulli E, Colafigli G, Pepe S, Lisi C, Marchetti P, Martelli M, Foà R, Breccia M. Management of myelofibrosis and concomitant advanced cutaneous squamous cell carcinoma with ruxolitinib associated with cemiplimab. Ann Hematol 2020; 100:2117-2119. [PMID: 32856142 DOI: 10.1007/s00277-020-04236-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/24/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Alessio Di Prima
- Hematology, Department of Translational and Precision Medicine, Azienda Ospedaliera Policlinico Umberto 1, Sapienza University, Via Benevento 6, 00161, Rome, Italy
| | - Andrea Botticelli
- Clinical and Molecular Department, Oncologia B Policlinico Umberto I, Sapienza University, Rome, Italy
| | - Emilia Scalzulli
- Hematology, Department of Translational and Precision Medicine, Azienda Ospedaliera Policlinico Umberto 1, Sapienza University, Via Benevento 6, 00161, Rome, Italy
| | - Gioia Colafigli
- Hematology, Department of Translational and Precision Medicine, Azienda Ospedaliera Policlinico Umberto 1, Sapienza University, Via Benevento 6, 00161, Rome, Italy
| | - Sara Pepe
- Hematology, Department of Translational and Precision Medicine, Azienda Ospedaliera Policlinico Umberto 1, Sapienza University, Via Benevento 6, 00161, Rome, Italy
| | - Chiara Lisi
- Hematology, Department of Translational and Precision Medicine, Azienda Ospedaliera Policlinico Umberto 1, Sapienza University, Via Benevento 6, 00161, Rome, Italy
| | - Paolo Marchetti
- Clinical and Molecular Department, Oncologia B Policlinico Umberto I, Sapienza University, Rome, Italy
| | - Maurizio Martelli
- Hematology, Department of Translational and Precision Medicine, Azienda Ospedaliera Policlinico Umberto 1, Sapienza University, Via Benevento 6, 00161, Rome, Italy
| | - Robin Foà
- Hematology, Department of Translational and Precision Medicine, Azienda Ospedaliera Policlinico Umberto 1, Sapienza University, Via Benevento 6, 00161, Rome, Italy
| | - Massimo Breccia
- Hematology, Department of Translational and Precision Medicine, Azienda Ospedaliera Policlinico Umberto 1, Sapienza University, Via Benevento 6, 00161, Rome, Italy.
| |
Collapse
|
26
|
Coricello A, Mesiti F, Lupia A, Maruca A, Alcaro S. Inside Perspective of the Synthetic and Computational Toolbox of JAK Inhibitors: Recent Updates. Molecules 2020; 25:E3321. [PMID: 32707925 PMCID: PMC7435994 DOI: 10.3390/molecules25153321] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 01/10/2023] Open
Abstract
The mechanisms of inflammation and cancer are intertwined by complex networks of signaling pathways. Dysregulations in the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway underlie several pathogenic conditions related to chronic inflammatory states, autoimmune diseases and cancer. Historically, the potential application of JAK inhibition has been thoroughly explored, thus triggering an escalation of favorable results in this field. So far, five JAK inhibitors have been approved by the Food and Drug Administration (FDA) for the treatment of different diseases. Considering the complexity of JAK-depending processes and their involvement in multiple disorders, JAK inhibitors are the perfect candidates for drug repurposing and for the assessment of multitarget strategies. Herein we reviewed the recent progress concerning JAK inhibition, including the innovations provided by the release of JAKs crystal structures and the improvement of synthetic strategies aimed to simplify of the industrial scale-up.
Collapse
Affiliation(s)
- Adriana Coricello
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Francesco Mesiti
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
- Net4Science srl, Università 'Magna Græcia' di Catanzaro, Campus Universitario 'S. Venuta', Viale Europa, 88100 Catanzaro, Italy
| | - Antonio Lupia
- Net4Science srl, Università 'Magna Græcia' di Catanzaro, Campus Universitario 'S. Venuta', Viale Europa, 88100 Catanzaro, Italy
| | - Annalisa Maruca
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
- Net4Science srl, Università 'Magna Græcia' di Catanzaro, Campus Universitario 'S. Venuta', Viale Europa, 88100 Catanzaro, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
- Net4Science srl, Università 'Magna Græcia' di Catanzaro, Campus Universitario 'S. Venuta', Viale Europa, 88100 Catanzaro, Italy
| |
Collapse
|
27
|
Li L, He X, Wang X, Sun Y, Wu G, Fang H, Wang C, Luo P, Xia Z. Ruxolitinib protects lipopolysaccharide (LPS)-induced sepsis through inhibition of nitric oxide production in mice. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:546. [PMID: 32411769 PMCID: PMC7214887 DOI: 10.21037/atm-20-2972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background Ruxolitinib is an inhibitor of Janus kinases (JAK) 1/2. It was authorised recently by the U.S. Food and Drug Administration (FDA) as a new Myelofibrosis treatment. In this study, we identified ruxolitinib as a new inhibitor of nitric oxide (NO) production in response to lipopolysaccharide (LPS)stimulation of RAW 264.7 cells. Methods In vitro direct effects of ruxolitinib were determined through NO production on RAW 264.7 cells. Also the expression level of iNOS, TNF-α and IL-6 were detected by Western Blotting and qRT-PCR. In vivo therapeutic effects of ruxolitinib on sepsis were evaluated by an endotoxemia model with C57 mice. The survival was calculated and histopathological damage of organs was observed by HE. Cytokines in serum were detected by Mouse Cytokine Array Panel. Results Ruxolitinib was found to significantly reduce NO production, inducible nitric oxide synthase (iNOS), TNF-α, and IL-6 expression, suggesting that ruxolitinib blocks LPS signaling that leads to pro-inflammatory factor expression. Furthermore, the inhibitory effects of ruxolitinib contributed to the survival of septic mice by 70% and pro-inflammatory cytokines in serum declined apparently. The results taken together indicate that ruxolitinib can significantly suppress LPS-stimulated NO production and improve the survival of septic mice, perhaps by interfering with the NF-κB pathway. Conclusions These findings suggest ruxolitinib might be a possible therapeutic candidate for sepsis therapy.
Collapse
Affiliation(s)
- Li Li
- Department of Burn Surgery, Affiliated Changhai Hospital of the Navy Medical University, Shanghai 200433, China
| | - Xingfeng He
- Department of Burn Surgery, Affiliated Changhai Hospital of the Navy Medical University, Shanghai 200433, China.,Department of Burns and Plastic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xingtong Wang
- Department of Burns and Plastic Surgery, The Sixth Medical Center of General Hospital, The People's Liberation Army, Beijing 100046, China
| | - Yu Sun
- Department of Burn Surgery, Affiliated Changhai Hospital of the Navy Medical University, Shanghai 200433, China
| | - Guosheng Wu
- Department of Burn Surgery, Affiliated Changhai Hospital of the Navy Medical University, Shanghai 200433, China
| | - He Fang
- Department of Burn Surgery, Affiliated Changhai Hospital of the Navy Medical University, Shanghai 200433, China
| | - Chen Wang
- Department of Burn Surgery, Affiliated Changhai Hospital of the Navy Medical University, Shanghai 200433, China
| | - Pengfei Luo
- Department of Burn Surgery, Affiliated Changhai Hospital of the Navy Medical University, Shanghai 200433, China
| | - Zhaofan Xia
- Department of Burn Surgery, Affiliated Changhai Hospital of the Navy Medical University, Shanghai 200433, China.,Department of Burns and Plastic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
28
|
Song X, Liu Z, Yu Z. EGFR Promotes the Development of Triple Negative Breast Cancer Through JAK/STAT3 Signaling. Cancer Manag Res 2020; 12:703-717. [PMID: 32099467 PMCID: PMC6996555 DOI: 10.2147/cmar.s225376] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/07/2019] [Indexed: 12/28/2022] Open
Abstract
Purpose To investigate the role of EGFR and STAT3 in breast cancer development and progression. Methods Through bioinformatics analysis differently expressed genes (DEGs) including EGFR and STAT3 were identified in breast cancer tissue. QRT-PCR and Western blot analysis were used to investigate EGFR and STAT3 levels in breast cancer tissues and cells. The influence of EGFR and STAT3 on the breast cancer cell proliferation (CCK-8 assay, clone formation assays), migration (wound healing assays) and invasion (transwell assays) were investigated. The influence of EGFR on breast cancer in vivo was examined by Nude mouse transplantation tumor experiments and immunohistochemistry (IHC) staining. The effects of EGFR on breast cancer signaling were assessed via Western blot. Results Both EGFR and p-STAT3 were up-regulated in breast cancer tissues and cell lines. EGFR expression was positively associated with p-STAT3. Moreover, EGFR and p-STAT3 activity enhanced the proliferation and invasion of tumor cells. Breast cancer cell growth was dramatically inhibited by EGFR silencing in vivo. Conclusion EGFR promotes breast cancer progression via STAT3 phosphorylation and JAK/STAT3 signaling.
Collapse
Affiliation(s)
- Xiang Song
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| | - Zhaoyun Liu
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China.,School of Medicine, Shandong University, Jinan, People's Republic of China
| | - Zhiyong Yu
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong, People's Republic of China
| |
Collapse
|
29
|
Breccia M, Baratè C, Benevolo G, Bonifacio M, Elli EM, Guglielmelli P, Maffioli M, Malato A, Mendicino F, Palumbo GA, Pugliese N, Rossi E, Rumi E, Sant'Antonio E, Ricco A, Tiribelli M, Palandri F. Tracing the decision-making process for myelofibrosis: diagnosis, stratification, and management of ruxolitinib therapy in real-word practice. Ann Hematol 2020; 99:65-72. [PMID: 31832751 PMCID: PMC6944647 DOI: 10.1007/s00277-019-03847-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/12/2019] [Indexed: 12/17/2022]
Abstract
The management of patients with myelofibrosis (MF) has dramatically changed since the introduction of ruxolitinib as a tailored treatment strategy. However, the perceptions about the use of this drug in clinical practice remain, at times, a matter of discussion. We conducted a survey about the diagnostic evaluation, prognostic assessment, and management of ruxolitinib in real-life clinical practice in 18 Italian hematology centers. At diagnosis, most hematologists do not use genetically or molecularly inspired score systems to assess prognosis, mainly due to scarce availability of next-generation sequencing (NGS) methodology, with NGS conversely reserved only for a subset of lower-risk MF patients with the aim of possibly improving the treatment strategy. Some common points in the management of ruxolitinib were 1) clinical triggers for ruxolitinib therapy, regardless of risk category; 2) evaluation of infectious risk before the starting of the drug; and 3) schedule of monitoring during the first 12 weeks with the need, in some instances, of supportive treatment. Further development of international recommendations and insights will allow the achievement of common criteria for the management of ruxolitinib in MF, before and after treatment, and for the definition of response and failure.
Collapse
Affiliation(s)
- Massimo Breccia
- Hematology, Department of Precision and Translational Medicine, Policlinico Umberto 1, Sapienza University, Rome, Italy.
| | - Claudia Baratè
- Department of Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy
| | - Giulia Benevolo
- Hematology, Città della Salute e della Scienza, Turin, Italy
| | | | - Elena Maria Elli
- Hematology Division and Bone Marrow Transplant Unit, ASST Monza, Ospedale San Gerardo, Monza, Italy
| | - Paola Guglielmelli
- Center of Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, University of Florence, Florence, Italy
| | | | - Alessandra Malato
- Division of Hematology and Bone Marrow Transplant, Villa Sofia-Cervello Hospital, 90146, Palermo, Italy
| | - Francesco Mendicino
- Hematology Unit, Department of Hemato-Oncology, Ospedale Annunziata, Via San Martino, snc -, 87100, Cosenza, Italy
| | - Giuseppe Alberto Palumbo
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie Avanzate "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Novella Pugliese
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Elena Rossi
- Istituto di Ematologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Elisa Rumi
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Emanuela Sant'Antonio
- Department of Oncology, Division of Hematology, Azienda USL Toscana Nord Ovest, Lucca, Italy
- Medical Genetics, University of Siena, Siena, Italy
| | - Alessandra Ricco
- Department of Emergency and Organ Transplantation (DETO), Hematology Section, University of Bari, Bari, Italy
| | - Mario Tiribelli
- Division of Hematology and Bone Marrow Transplantation, Department of Medical Area, University of Udine, Udine, Italy
| | - Francesca Palandri
- Institute of Hematology "L. and A. Seràgnoli", Sant'Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
30
|
Bai Y, Wang W, Yin P, Gao J, Na L, Sun Y, Wang Z, Zhang Z, Zhao C. Ruxolitinib Alleviates Renal Interstitial Fibrosis in UUO Mice. Int J Biol Sci 2020; 16:194-203. [PMID: 31929748 PMCID: PMC6949153 DOI: 10.7150/ijbs.39024] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023] Open
Abstract
Ruxolitinib is a selective inhibitor of Jak1/2. Downstream signaling pathways of Jak, such as Stat3 and Akt/mTOR, are overactivated and contribute to renal interstitial fibrosis. Therefore, we explored the effect of Ruxolitinib on this pathological process. Unilateral ureteral obstruction (UUO) models and TGF-β1-treated fibroblasts and renal tubular epithelial cells were adopted in this study. Ruxolitinib was administered to UUO mice and TGF-β1-treated cells. Kidneys from UUO mice with Ruxolitinib treatment displayed less tubular injuries compared with those without Ruxolitinib treatment. Ruxolitinib treatment suppressed fibroblast activation and extracellular matrix (ECM) production in UUO kidneys and TGF-β1-treated fibroblasts. Ruxolitinib treatment also blocked epithelial-mesenchymal transition (EMT) in UUO kidneys and TGF-β 1-treated renal tubular epithelial cells. Moreover, Ruxolitinib treatment alleviated UUO-induced inflammation, oxidative stress and apoptosis. Mechanistically, Ruxolitinib treatment attenuated activation of both Stat3 and Akt/mTOR/Yap pathways. In conclusion, Ruxolitinib treatment can ameliorate UUO-induced renal interstitial fibrosis, suggesting that Ruxolitinib may be potentially used to treat fibrotic kidney disease.
Collapse
Affiliation(s)
- Yu Bai
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China.,Department of Nephrology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Ping Yin
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Jian Gao
- Center of Laboratory Technology and Experimental Medicine, China Medical University, Shenyang, China
| | - Lei Na
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yu Sun
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Zhuo Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Zhongbo Zhang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Chenghai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
31
|
Yang T, Hu M, Qi W, Yang Z, Tang M, He J, Chen Y, Bai P, Yuan X, Zhang C, Liu K, Lu Y, Xiang M, Chen L. Discovery of Potent and Orally Effective Dual Janus Kinase 2/FLT3 Inhibitors for the Treatment of Acute Myelogenous Leukemia and Myeloproliferative Neoplasms. J Med Chem 2019; 62:10305-10320. [PMID: 31670517 DOI: 10.1021/acs.jmedchem.9b01348] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Tao Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Mengshi Hu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenyan Qi
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Jun He
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yong Chen
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Peng Bai
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Xue Yuan
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Chufeng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Kongjun Liu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yulin Lu
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Mingli Xiang
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Lijuan Chen
- State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
32
|
Parra-Izquierdo I, Castaños-Mollor I, López J, Gómez C, San Román JA, Sánchez Crespo M, García-Rodríguez C. Lipopolysaccharide and interferon-γ team up to activate HIF-1α via STAT1 in normoxia and exhibit sex differences in human aortic valve interstitial cells. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2168-2179. [PMID: 31034990 DOI: 10.1016/j.bbadis.2019.04.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/17/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022]
Abstract
In early stages of calcific aortic valve disease (CAVD), immune cells infiltrate into the valve leaflets and release cytokines such as interferon (IFN)-γ. IFN-γ has context-dependent direct effects, and also regulates other immune pathways. The purpose of this study was addressing the effects of IFN-γ on human aortic valve interstitial cells (AVICs), focusing on the pathogenic processes underlying CAVD. Strikingly, under normoxic conditions, IFN-γ induced hypoxia inducible factor (HIF)-1α expression, an effect strongly potentiated by the Toll-like receptor (TLR)-4 ligand lipopolysaccharide (LPS). Immunodetection studies confirmed the nuclear translocation of HIF-1α. Gene silencing showed that HIF-1α expression is dependent on signal transducer and activator of transcription (STAT)-1 expression. Consistent with HIF-1α induction, the secretion of the endothelial growth factor was detected by ELISA, and downregulation of the antiangiogenic factor chondromodulin-1 gene was observed by qPCR. Results also disclosed IFN-γ as a proinflammatory cytokine that cooperates with LPS to induce the expression of adhesion molecules, prostaglandin E2 and interleukins. Moreover, IFN-γ induced an osteogenic phenotype and promoted in vitro calcification that were markedly potentiated by LPS. Pharmacological experiments disclosed the involvement of Janus Kinases (JAK)/STATs as well as ERK/HIF-1α routes on the induction of calcification. Notably, IFN-γ receptor 1 expression, as well as ERK/HIF-1α activation, and the subsequent responses were more robust in male AVICs. This is the first report uncovering an immune and non-hypoxic activation of HIF-1α via STAT1 in AVIC. The aforementioned results and the sex-differential responses may be potentially relevant to better understand CAVD pathogenesis.
Collapse
Affiliation(s)
- Iván Parra-Izquierdo
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Irene Castaños-Mollor
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Javier López
- ICICOR, Hospital Clínico Universitario, Valladolid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Cristina Gómez
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| | - J Alberto San Román
- ICICOR, Hospital Clínico Universitario, Valladolid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Mariano Sánchez Crespo
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Carmen García-Rodríguez
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Spain.
| |
Collapse
|
33
|
Barrett LE, Gardner HL, Barber LG, Sadowski A, London CA. Safety and toxicity of combined oclacitinib and carboplatin or doxorubicin in dogs with solid tumors: a pilot study. BMC Vet Res 2019; 15:291. [PMID: 31409327 PMCID: PMC6693187 DOI: 10.1186/s12917-019-2032-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 07/30/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Oclacitinib is an orally bioavailable Janus Kinase (JAK) inhibitor approved for the treatment of canine atopic dermatitis. Aberrant JAK/ Signal Transducer and Activator of Transcription (STAT) signaling within hematologic and solid tumors has been implicated as a driver of tumor growth through effects on the local microenvironment, enhancing angiogenesis, immune suppression, among others. A combination of JAK/STAT inhibition with cytotoxic chemotherapy may therefore result in synergistic anti-cancer activity, however there is concern for enhanced toxicities. The purpose of this study was to evaluate the safety profile of oclacitinib given in combination with either carboplatin or doxorubicin in tumor-bearing dogs. RESULT Oclacitinib was administered at the label dose of 0.4-0.6 mg/kg PO q12h in combination with either carboplatin at 250-300 mg/m2 or doxorubicin at 30 mg/m2 IV q21d. Nine dogs were enrolled in this pilot study (n = 4 carboplatin; n = 5 doxorubicin). No unexpected toxicities occurred, and the incidence of adverse events with combination therapy was not increased beyond that expected in dogs treated with single agent chemotherapy. Serious adverse events included one Grade 4 thrombocytopenia and one Grade 4 neutropenia. No objective responses were noted. CONCLUSIONS Oclacitinib is well tolerated when given in combination with carboplatin or doxorubicin. Future work is needed to explore whether efficacy is enhanced in this setting.
Collapse
Affiliation(s)
- Laura E Barrett
- Cummings School, Tufts University, Foster Hospital for Small Animals, 200 Westboro Rd, N., Grafton, MA, 01536, USA
| | - Heather L Gardner
- Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, USA
| | - Lisa G Barber
- Cummings School, Tufts University, Foster Hospital for Small Animals, 200 Westboro Rd, N., Grafton, MA, 01536, USA
| | - Abbey Sadowski
- Cummings School, Tufts University, Foster Hospital for Small Animals, 200 Westboro Rd, N., Grafton, MA, 01536, USA
| | - Cheryl A London
- Cummings School, Tufts University, Foster Hospital for Small Animals, 200 Westboro Rd, N., Grafton, MA, 01536, USA.
| |
Collapse
|
34
|
Chang WM, Chang YC, Yang YC, Lin SK, Chang PMH, Hsiao M. AKR1C1 controls cisplatin-resistance in head and neck squamous cell carcinoma through cross-talk with the STAT1/3 signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:245. [PMID: 31182137 PMCID: PMC6558898 DOI: 10.1186/s13046-019-1256-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/30/2019] [Indexed: 01/22/2023]
Abstract
Background Cisplatin is the first-line chemotherapy used against most upper aerodigestive tract carcinomas. In head and neck cancer, sensitivity to cisplatin remains the key issue in treatment response and outcome. Genetic heterogeneity and aberrant gene expression may be the intrinsic factors that cause primary cisplatin-resistance. Methods Combination of the HNSCC gene expression data and the cisplatin sensitivity results from public database. We found that aldo-keto reductase family 1 member C1 (AKR1C1) may be associated with cisplatin sensitivity in HNSCC treatment of naïve cells. We examined the AKR1C1 expression and its correlation with cisplatin IC50 and prognosis in patients. The in vitro and in vivo AKR1C1 functions in cisplatin-resistance through overexpression or knockdown assays, respectively. cDNA microarrays were used to identify the upstream regulators that modulate AKR1C1-induced signaling in HNSCC. Finally, we used the cigarette metabolites to promote AKR1C1 expression and ruxolitinib to overcome AKR1C1-induced cisplatin-resistance. Results AKR1C1 positively correlates to cisplatin-resistance in HNSCC cells. AKR1C1 is a poor prognostic factor for recurrence and death of HNSCC patients. Silencing of AKR1C1 not only reduced in vitro IC50 but also increased in vivo cisplatin responses and vise versa in overexpression cells. Cigarette metabolites also promote AKR1C1 expression. Transcriptome analyses revealed that STAT1 and STAT3 activation enable AKR1C1-induced cisplatin-resistance and can be overcome by ruxolitinib treatment. Conclusions AKR1C1 is a crucial regulator for cisplatin-resistance in HNSCC and also poor prognostic marker for patients. Targeting the AKR1C1-STAT axis may provide a new therapeutic strategy to treat patients who are refractory to cisplatin treatment. Electronic supplementary material The online version of this article (10.1186/s13046-019-1256-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei-Min Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chan Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Chieh Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Sze-Kwan Lin
- Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Peter Mu-Hsin Chang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan. .,Faculty of Medicine, College of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan. .,Department of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
35
|
Roskoski R. Properties of FDA-approved small molecule protein kinase inhibitors. Pharmacol Res 2019; 144:19-50. [DOI: 10.1016/j.phrs.2019.03.006] [Citation(s) in RCA: 360] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 12/14/2022]
|
36
|
Abstract
Atopic dermatitis (AD) is one of the most common inflammatory skin diseases. AD is driven by barrier dysfunction and abnormal immune activation of T helper (Th) 2, Th22, and varying degrees of Th1 and Th17 among various subtypes. The Janus kinase (JAK)-signal transducer and activator of transcription (STAT) and spleen tyrosine kinase (SYK) pathways are involved in signaling of several AD-related cytokines, such as IFN-γ, IL-4, IL-13, IL-31, IL-33, IL-23, IL-22, and IL-17, mediating downstream inflammation and barrier alterations. While AD is primarily Th2-driven, the clinical and molecular heterogeneity of AD endotypes highlights the unmet need for effective therapeutic options that target more than one immune axis and are safe for long-term use. The JAK inhibitors, which target different combinations of kinases, have overlapping but distinct mechanisms of action and safety profiles. Several topical and oral JAK inhibitors have been shown to decrease AD severity and symptoms. A review of the JAK and SYK inhibitors that are currently undergoing evaluation for efficacy and safety in the treatment of AD summarizes available data on a promising area of therapeutics and further elucidates the complex molecular interactions that drive AD.
Collapse
Affiliation(s)
- Helen He
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, 5 E. 98th Street, New York, NY, 10029, USA
| | - Emma Guttman-Yassky
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, 5 E. 98th Street, New York, NY, 10029, USA.
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
37
|
Hu M, Xu C, Yang C, Zuo H, Chen C, Zhang D, Shi G, Wang W, Shi J, Zhang T. Discovery and evaluation of ZT55, a novel highly-selective tyrosine kinase inhibitor of JAK2 V617F against myeloproliferative neoplasms. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:49. [PMID: 30717771 PMCID: PMC6360668 DOI: 10.1186/s13046-019-1062-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/27/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND The JAK2-STAT signaling pathway plays a critical role in myeloproliferative neoplasms (MPN). An activating mutation in JAK2 (V617F) is present in ~ 95% of polycythemia vera, essential thrombocythemia, and primary myelofibrosis cases. This study aims to explore the selective JAK2V617F inhibitor, evaluate the efficacy and possible mechanism of ZT55 on MPN. METHODS HTRF assays were conducted to evaluate the selective inhibition of ZT55 for JAKs. Cell apoptosis, proliferation, and cycle arrest assays were performed to examine the effect of ZT55 on HEL cell line with JAK2V617F mutation in vitro. Western analysis was used to monitor the expression and activity of proteins on JAK2/STAT pathway. A mice xenograft model was established to evaluate the antitumor efficacy of ZT55 in vivo. Peripheral blood samples from patients with the JAK2V617F mutation were collected to estimate the effect of ZT55 on erythroid colony formation by colony-forming assay. RESULTS We found that ZT55 showed a selective inhibition of a 0.031 μM IC50 value against JAK2. It exhibited potent effects on the cellular JAK-STAT pathway, inhibiting tyrosine phosphorylation in JAK2V617F and downstream STAT3/5 transcription factors. ZT55 inhibited the proliferation of the JAK2V617F-expressing HEL cell line, leading to cell cycle arrest at the G2/M phase and induction of caspase-dependent apoptosis. Notably, ZT55 also significantly suppressed the growth of HEL xenograft tumors in vivo. Further evaluation indicated that ZT55 blocked erythroid colony formation of peripheral blood hematopoietic progenitors from patients carrying the JAK2V617F mutation. CONCLUSION These results suggest that ZT55 is a highly-selective JAK2 inhibitor that can induce apoptosis of human erythroleukemia cells by inhibiting the JAK2-STAT signaling.
Collapse
Affiliation(s)
- Min Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Chengbo Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Chao Yang
- Department of Blood Transfusion, General Hospital of the PLA Rocket Force, Beijing, 100088, China
| | - Hongli Zuo
- Department of Hematology, 307 Hospital of the PLA, Beijing, 100071, China
| | - Chengjuan Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Dan Zhang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Gaona Shi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Wenjie Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jiangong Shi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China. .,School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China.
| |
Collapse
|
38
|
Wiedemann B, Weisner J, Rauh D. Chemical modulation of transcription factors. MEDCHEMCOMM 2018; 9:1249-1272. [PMID: 30151079 PMCID: PMC6097187 DOI: 10.1039/c8md00273h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
Transcription factors (TFs) constitute a diverse class of sequence-specific DNA-binding proteins, which are key to the modulation of gene expression. TFs have been associated with human diseases, including cancer, Alzheimer's and other neurodegenerative diseases, which makes this class of proteins attractive targets for chemical biology and medicinal chemistry research. Since TFs lack a common binding site or structural similarity, the development of small molecules to efficiently modulate TF biology in cells and in vivo is a challenging task. This review highlights various strategies that are currently being explored for the identification and development of modulators of Myc, p53, Stat, Nrf2, CREB, ER, AR, HIF, NF-κB, and BET proteins.
Collapse
Affiliation(s)
- Bianca Wiedemann
- Technische Universität Dortmund , Fakultät für Chemie und Chemische Biologie , Otto-Hahn-Strasse 4a , D-44227 Dortmund , Germany . ; ; Tel: +49 (0)231 755 7080
| | - Jörn Weisner
- Technische Universität Dortmund , Fakultät für Chemie und Chemische Biologie , Otto-Hahn-Strasse 4a , D-44227 Dortmund , Germany . ; ; Tel: +49 (0)231 755 7080
| | - Daniel Rauh
- Technische Universität Dortmund , Fakultät für Chemie und Chemische Biologie , Otto-Hahn-Strasse 4a , D-44227 Dortmund , Germany . ; ; Tel: +49 (0)231 755 7080
| |
Collapse
|
39
|
Huang Y, Dong G, Li H, Liu N, Zhang W, Sheng C. Discovery of Janus Kinase 2 (JAK2) and Histone Deacetylase (HDAC) Dual Inhibitors as a Novel Strategy for the Combinational Treatment of Leukemia and Invasive Fungal Infections. J Med Chem 2018; 61:6056-6074. [PMID: 29940115 DOI: 10.1021/acs.jmedchem.8b00393] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Clinically, leukemia patients often suffer from the limited efficacy of chemotherapy and high risks of infection by invasive fungal pathogens. Herein, a novel therapeutic strategy was developed in which a small molecule can simultaneously treat leukemia and invasive fungal infections (IFIs). Novel Janus kinase 2 (JAK2) and histone deacetylase (HDAC) dual inhibitors were identified to possess potent anti-proliferative activity toward hematological cell lines and excellent synergistic effects with fluconazole to treat resistant Candida albicans infections. In particular, compound 20a, a highly active and selective JAK2/HDAC6 dual inhibitor, showed excellent in vivo antitumor efficacy in several acute myeloid leukemia (AML) models and synergized with fluconazole for the treatment of resistant C. albicans infections. This study highlights the therapeutic potential of JAK2/HDAC dual inhibitors in treating AML and IFIs and provides an efficient strategy for multitargeting drug discovery.
Collapse
Affiliation(s)
- Yahui Huang
- School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , PR China
| | - Guoqiang Dong
- School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , PR China
| | - Huanqiu Li
- College of Pharmaceutical Science , Soochow University , Suzhou 215123 , PR China
| | - Na Liu
- School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , PR China
| | - Wannian Zhang
- School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , PR China
| | - Chunquan Sheng
- School of Pharmacy , Second Military Medical University , 325 Guohe Road , Shanghai 200433 , PR China
| |
Collapse
|
40
|
Pogorzelska A, Sławiński J, Kawiak A, Żołnowska B, Chojnacki J, Stasiłojć G, Ulenberg S, Szafrański K, Bączek T. Synthesis, molecular structure, and metabolic stability of new series of N'-(2-alkylthio-4-chloro-5-methylbenzenesulfonyl)-1-(5-phenyl-1H-pyrazol-1-yl)amidine as potential anti-cancer agents. Eur J Med Chem 2018; 155:670-680. [PMID: 29936354 DOI: 10.1016/j.ejmech.2018.06.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/28/2018] [Accepted: 06/12/2018] [Indexed: 11/26/2022]
Abstract
A series of new N'-(2-alkylthio-4-chloro-5-methylbenzenesulfonyl)-1-(5-phenyl-1H-pyrazol-1-yl)amidine derivatives have been synthesized and evaluated in vitro by MTT assays for their antiproliferative activity against cell lines of colon cancer HCT-116, cervical cancer HeLa and breast cancer MCF-7. The studied compounds display selective activity mainly against HCT-116 and HeLa cells. Thus, five compounds show selective cytotoxic effect against HCT-116 (IC50 = 3-10 μM) and HeLa (IC50 = 7 μM). Importantly, the noticed values of IC50 for four compounds are almost 4-fold lower for HeLa than non-malignant HaCaT cells. More-in-depth biological research revealed that the treatment of HCT-116 and HeLa with active compound resulted in increased numbers of cells in sub-G1 phase in a time dependent manner, while non-active derivative does not influence cell cycle. Metabolic stability assays using liver microsomes and NADPH provide important information on compounds susceptibility to phase 1 biotransformation reactions.
Collapse
Affiliation(s)
- Aneta Pogorzelska
- Department of Organic Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416, Gdańsk, Poland.
| | - Jarosław Sławiński
- Department of Organic Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416, Gdańsk, Poland.
| | - Anna Kawiak
- Department of Biotechnology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Ul. Abrahama 58, 80-307, Gdańsk, Poland; Laboratory of Human Physiology, Medical University of Gdańsk, Ul. Tuwima 15, 80-210, Gdańsk, Poland
| | - Beata Żołnowska
- Department of Organic Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416, Gdańsk, Poland
| | - Jarosław Chojnacki
- Department of Inorganic Chemistry, Gdańsk University of Technology, Narutowicza 11/12, 80-233, Gdańsk, Poland
| | - Grzegorz Stasiłojć
- Laboratory of Cell Biology, Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdańsk, Ul. Dębinki 1, Gdańsk, 80-211, Poland
| | - Szymon Ulenberg
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416, Gdańsk, Poland
| | - Krzysztof Szafrański
- Department of Organic Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416, Gdańsk, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416, Gdańsk, Poland
| |
Collapse
|
41
|
Aboul-Fettouh N, Nijhawan RI. Aggressive squamous cell carcinoma in a patient on the Janus kinase inhibitor ruxolitinib. JAAD Case Rep 2018; 4:455-457. [PMID: 29984281 PMCID: PMC6031591 DOI: 10.1016/j.jdcr.2018.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Nader Aboul-Fettouh
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rajiv I Nijhawan
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
42
|
Turbeville JG, Patel NU, Cardwell LA, Oussedik E, Feldman SR. Recent Advances in Small Molecule and Biological Therapeutic Approaches in the Treatment of Psoriasis. Clin Pharmacol Ther 2017; 102:70-85. [DOI: 10.1002/cpt.688] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/10/2017] [Accepted: 03/11/2017] [Indexed: 01/31/2023]
Affiliation(s)
- JG Turbeville
- Center for Dermatology Research; Department of Dermatology, Wake Forest School of Medicine; Winston-Salem North Carolina USA
| | - NU Patel
- Center for Dermatology Research; Department of Dermatology, Wake Forest School of Medicine; Winston-Salem North Carolina USA
| | - LA Cardwell
- Center for Dermatology Research; Department of Dermatology, Wake Forest School of Medicine; Winston-Salem North Carolina USA
| | - E Oussedik
- Center for Dermatology Research; Department of Dermatology, Wake Forest School of Medicine; Winston-Salem North Carolina USA
| | - SR Feldman
- Center for Dermatology Research; Department of Dermatology, Wake Forest School of Medicine; Winston-Salem North Carolina USA
- Department of Pathology; Wake Forest School of Medicine; Winston-Salem North Carolina USA
- Department of Public Health Sciences; Wake Forest School of Medicine; Winston-Salem North Carolina USA
| |
Collapse
|
43
|
Alboini PE, Evoli A, Damato V, Iorio R, Bartoccioni E. Remission of myasthenia gravis with MuSK antibodies during ruxolitinib treatment. Muscle Nerve 2016; 55:E12-E13. [DOI: 10.1002/mus.25458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 10/25/2016] [Accepted: 10/28/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Paolo E. Alboini
- Institute of Neurology; Largo F. Vito, 1 - 00168, Catholic University Rome Italy
| | - Amelia Evoli
- Institute of Neurology; Largo F. Vito, 1 - 00168, Catholic University Rome Italy
| | - Valentina Damato
- Institute of Neurology; Largo F. Vito, 1 - 00168, Catholic University Rome Italy
| | - Raffaele Iorio
- Institute of Neurology; Largo F. Vito, 1 - 00168, Catholic University Rome Italy
| | | |
Collapse
|
44
|
Smaili W, Doubaj Y, Laarabi FZ, Lyahyai J, Kerbout M, Mikdame M, Sefiani A. CALR gene mutational profile in myeloproliferative neoplasms with non-mutated JAK2 in Moroccan patients: A case series and germline in-frame deletion. Curr Res Transl Med 2016; 65:15-19. [PMID: 28340692 DOI: 10.1016/j.retram.2016.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/05/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND The discovery of somatic mutations within the gene encoding calreticulin (CALR) in 2013 represented a major milestone in the molecular diagnosis of BCR-ABL negative myeloproliferative neoplasms (MPN). In fact, exome sequencing revealed that most patients with essential thrombocythemia (ET) or primary myelofibrosis (PMF) lacking JAK2 or MPL mutations, harbor somatic insertion and/or deletion in exon 9 of CALR gene. In this study, we identified the first CALR gene mutational landscape in Moroccan patients with MPN nonmutated for the JAK2 gene. METHODS We performed Sanger sequencing of exon 9 of CALR gene in blood samples obtained from 33 Moroccan patients with ET or PMF non-mutated for JAK2. RESULTS Of the 33 patients analyzed, we detected eight distinct variants in 15 patients (45.4%); six indel mutations, five with type 1 recurrent 52bp deletion, four with type 2 recurrent 5bp insertion and one in frame deletion which was found to be a germline variant suggesting a very rare condition in MPN. CONCLUSION This is the first cohort reported in CALR gene mutation analysis in Morocco. Our results were concordant with studies reported up to date and very encouraging in promoting the molecular diagnosis of myeloproliferative neoplasms in Moroccan patients. Moreover, the presence of a germline in frame deletion in a symptomatic patient should undermine the effectiveness of sizing assays without DNA sequencing in the diagnosis of CALR mutations.
Collapse
Affiliation(s)
- W Smaili
- Centre de génomique humaine, faculté de médecine et de pharmacie, université Mohamed V - Rabat, Rabat, Morocco; Département de génétique médicale, Institut national d'hygiène, 27, avenue Ibn Batouta, BP 769, 11400 Rabat, Morocco.
| | - Y Doubaj
- Centre de génomique humaine, faculté de médecine et de pharmacie, université Mohamed V - Rabat, Rabat, Morocco; Département de génétique médicale, Institut national d'hygiène, 27, avenue Ibn Batouta, BP 769, 11400 Rabat, Morocco
| | - F Z Laarabi
- Département de génétique médicale, Institut national d'hygiène, 27, avenue Ibn Batouta, BP 769, 11400 Rabat, Morocco
| | - J Lyahyai
- Centre de génomique humaine, faculté de médecine et de pharmacie, université Mohamed V - Rabat, Rabat, Morocco
| | - M Kerbout
- Service d'hématologie clinique, hôpital militaire d'instruction Mohammed V, université Mohamed V, Rabat, Morocco
| | - M Mikdame
- Service d'hématologie clinique, hôpital militaire d'instruction Mohammed V, université Mohamed V, Rabat, Morocco
| | - A Sefiani
- Centre de génomique humaine, faculté de médecine et de pharmacie, université Mohamed V - Rabat, Rabat, Morocco; Département de génétique médicale, Institut national d'hygiène, 27, avenue Ibn Batouta, BP 769, 11400 Rabat, Morocco
| |
Collapse
|
45
|
Abstract
THE ROLE OF RUXOLITINIB IN THE TREATMENT OF MYELOPROLIFERATIVE NEOPLASMS: The discovery of the JAK2V617F mutation in 2005, present in 95% of polycythemia vera (PV) and in 55% of myelofibrosis (MF) patients, opened the way for a new era of targeted therapies for myeloproliferative neoplasms. Ruxolitinib was the first-in-class Janus Kinase (JAK) inhibitor approved for the management of these diseases. In PV patients, conventional treatment strategies including aspirin, phlebotomy, cytoreductive agents such as hydroxyurea and interferon, clearly provide clinical benefits. However, some patients develop resistance or intolerance to these treatments. Ruxolitinib has been approved for PV patients who are resistant to or intolerant of hydroxyurea, based on the results of the phase 3 RESPONSE study. This study showed that ruxolitinib improves hematocrit control, reduces splenomegaly, and ameliorate disease-related symptoms as compared with best available therapy. In MF patients, the only curative treatment is allogeneic stem cell transplantation, but it remains restricted to a limited group of patients with poor prognosis and who are eligible for such procedure associated with non-negligible transplant-related mortality. Other treatments are palliative and unlikely to prolong survival. Ruxolitinib has been approved in the United States for MF patients with intermediate or high-risk disease, and in Europe for disease-related splenomegaly or symptoms in adults with MF, based on phase 3 COMFORT-I and COMFORT-II studies. These studies showed that ruxolitinib was able to reduce splenomegaly, ameliorate symptoms, and improve survival. However, the journey is not finished yet since there are still important unmet needs for MF patients, including improvement in cytopenias, and significant modification of disease natural history.
Collapse
Affiliation(s)
- Juliette Soret
- Centre d'Investigations Cliniques, Hôpital Saint-Louis, APHP, Paris, France
| | - Jean-Jacques Kiladjian
- Centre d'Investigations Cliniques, Hôpital Saint-Louis, APHP, Paris, France.; Inserm UMR-S 1131, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France.
| |
Collapse
|
46
|
Sullivan KD, Lewis HC, Hill AA, Pandey A, Jackson LP, Cabral JM, Smith KP, Liggett LA, Gomez EB, Galbraith MD, DeGregori J, Espinosa JM. Trisomy 21 consistently activates the interferon response. eLife 2016; 5:e16220. [PMID: 27472900 PMCID: PMC5012864 DOI: 10.7554/elife.16220] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 07/28/2016] [Indexed: 12/12/2022] Open
Abstract
Although it is clear that trisomy 21 causes Down syndrome, the molecular events acting downstream of the trisomy remain ill defined. Using complementary genomics analyses, we identified the interferon pathway as the major signaling cascade consistently activated by trisomy 21 in human cells. Transcriptome analysis revealed that trisomy 21 activates the interferon transcriptional response in fibroblast and lymphoblastoid cell lines, as well as circulating monocytes and T cells. Trisomy 21 cells show increased induction of interferon-stimulated genes and decreased expression of ribosomal proteins and translation factors. An shRNA screen determined that the interferon-activated kinases JAK1 and TYK2 suppress proliferation of trisomy 21 fibroblasts, and this defect is rescued by pharmacological JAK inhibition. Therefore, we propose that interferon activation, likely via increased gene dosage of the four interferon receptors encoded on chromosome 21, contributes to many of the clinical impacts of trisomy 21, and that interferon antagonists could have therapeutic benefits.
Collapse
Affiliation(s)
- Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Hannah C Lewis
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
| | - Amanda A Hill
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
| | - Ahwan Pandey
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Leisa P Jackson
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Joseph M Cabral
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
| | - L Alexander Liggett
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, United States
| | - Eliana B Gomez
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| | - James DeGregori
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, United States
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, United States
- Integrated Department of Immunology, University of Colorado School of Medicine, Aurora, United States
- Section of Hematology, University of Colorado School of Medicine, Aurora, United States
- Department of Medicine, University of Colorado School of Medicine, Aurora, United States
| | - Joaquín M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, United States
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, United States
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, United States
- Howard Hughes Medical Institute, Chevy Chase, United States
| |
Collapse
|
47
|
Tavallai M, Booth L, Roberts JL, Poklepovic A, Dent P. Rationally Repurposing Ruxolitinib (Jakafi (®)) as a Solid Tumor Therapeutic. Front Oncol 2016; 6:142. [PMID: 27379204 PMCID: PMC4904019 DOI: 10.3389/fonc.2016.00142] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 05/26/2016] [Indexed: 12/25/2022] Open
Abstract
We determined whether the approved myelofibrosis drug ruxolitinib (Jakafi®), an inhibitor of Janus kinases 1/2 (JAK1 and JAK2), could be repurposed as an anti-cancer agent for solid tumors. Ruxolitinib synergistically interacted with dual ERBB1/2/4 inhibitors to kill breast as well as lung, ovarian and brain cancer cells. Knock down of JAK1/2 or of ERBB1/2/3/4 recapitulated on-target drug effects. The combination of (ruxolitinib + ERBB1/2/4 inhibitor) rapidly inactivated AKT, mTORC1, mTORC2, STAT3, and STAT5, and activated eIF2α. In parallel, the drug combination reduced expression of MCL-1, BCL-XL, HSP90, HSP70, and GRP78, and increased expression of Beclin1. Activated forms of STAT3, AKT, or mTOR prevented the drug-induced decline in BCL-XL, MCL-1, HSP90, and HSP70 levels. Over-expression of chaperones maintained AKT/mTOR activity in the presence of drugs and protected tumor cells from the drug combination. Expression of dominant negative eIF2α S51A prevented the increase in Beclin1 expression and protected tumor cells from the drug combination. Loss of mTOR activity was associated with increased ATG13 S318 phosphorylation and with autophagosome formation. Autophagosomes initially co-localized with mitochondria and subsequently with lysosomes. Knock down of Beclin1 suppressed: drug-induced mitophagy; the activation of the toxic BH3 domain proteins BAX and BAK; and tumor cell killing. Knock down of apoptosis-inducing factor (AIF) protected tumor cells from the drug combination, whereas blockade of caspase 9 signaling did not. The drug combination released AIF into the cytosol and increased nuclear AIF: eIF3A co-localization. A 4-day transient exposure of orthotopic tumors to (ruxolitinib + afatinib) profoundly reduced mammary tumor growth over the following 35 days. Re-grown tumors exhibited high levels of BAD S112 phosphorylation and activation of ERK1/2 and NFκB. Our data demonstrate that mitophagy is an essential component of (ruxolitinib + ERBB inhibitor) lethality and that this drug combination should be explored in a phase I trial in solid tumor patients.
Collapse
Affiliation(s)
- Mehrad Tavallai
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University , Richmond, VA , USA
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University , Richmond, VA , USA
| | - Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University , Richmond, VA , USA
| | - Andrew Poklepovic
- Department of Medicine, Virginia Commonwealth University , Richmond, VA , USA
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University , Richmond, VA , USA
| |
Collapse
|
48
|
Reinwald M, Boch T, Hofmann WK, Buchheidt D. Risk of Infectious Complications in Hemato-Oncological Patients Treated with Kinase Inhibitors. Biomark Insights 2016; 10:55-68. [PMID: 27127405 PMCID: PMC4841329 DOI: 10.4137/bmi.s22430] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 09/13/2015] [Accepted: 09/15/2015] [Indexed: 12/30/2022] Open
Abstract
Infectious complications are a major cause of morbidity and mortality in patients with hemato-oncological diseases. Although disease-related immunosuppression represents one factor, aggressive treatment regimens, such as chemotherapy, stem cell transplantation, or antibody treatment, account for a large proportion of infectious side effects. With the advent of targeted therapies affecting specific kinases in malignant diseases, the outcome of patients has further improved. Nonetheless, dependent on the specific pathway targeted or off-target activity of the kinase inhibitor, therapy-associated infectious complications may occur. We review the most common and approved kinase inhibitors targeting a variety of hemato-oncological malignancies for their immunosuppressive potential and evaluate their risk of infectious side effects based on preclinical evidence and clinical data in order to raise awareness of the potential risks involved.
Collapse
Affiliation(s)
- Mark Reinwald
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Tobias Boch
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Dieter Buchheidt
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
49
|
Tavallai M, Booth L, Roberts JL, McGuire WP, Poklepovic A, Dent P. Ruxolitinib synergizes with DMF to kill via BIM+BAD-induced mitochondrial dysfunction and via reduced SOD2/TRX expression and ROS. Oncotarget 2016; 7:17290-300. [PMID: 26981780 PMCID: PMC4951212 DOI: 10.18632/oncotarget.8039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 02/29/2016] [Indexed: 01/01/2023] Open
Abstract
We determined whether the myelofibrosis drug ruxolitinib, an inhibitor of Janus kinases 1/2 (JAK1 and JAK2), could interact with the multiple sclerosis drug dimethyl-fumarate (DMF) to kill tumor cells; studies used the in vivo active form of the drug, mono-methyl fumarate (MMF). Ruxolitinib interacted with MMF to kill brain, breast, lung and ovarian cancer cells, and enhanced the lethality of standard of care therapies such as paclitaxel and temozolomide. MMF also interacted with other FDA approved drugs to kill tumor cells including Celebrex® and Gilenya®. The combination of [ruxolitinib + MMF] inactivated ERK1/2, AKT, STAT3 and STAT5; reduced expression of MCL-1, BCL-XL, SOD2 and TRX; increased BIM expression; decreased BAD S112 S136 phosphorylation; and enhanced pro-caspase 3 cleavage. Expression of activated forms of STAT3, MEK1 or AKT each significantly reduced drug combination lethality; prevented BAD S112 S136 dephosphorylation and decreased BIM expression; and preserved TRX, SOD2, MCL-1 and BCL-XL expression. The drug combination increased the levels of reactive oxygen species in cells, and over-expression of TRX or SOD2 prevented drug combination tumor cell killing. Over-expression of BCL-XL or knock down of BAX, BIM, BAD or apoptosis inducing factor (AIF) protected tumor cells. The drug combination increased AIF : HSP70 co-localization in the cytosol but this event did not prevent AIF : eIF3A association in the nucleus.
Collapse
Affiliation(s)
- Mehrad Tavallai
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Jane L. Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - William P. McGuire
- Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew Poklepovic
- Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
50
|
Shuly Y, Nagar M, Ben-Asaf L, Kneller A, Steinberg DM, Amariglio N, Salomon O. Calreticulin mutation burden--is it a stable clone in patients with essential thrombocythemia and myelofibrosis? Blood Cells Mol Dis 2015; 55:281-3. [PMID: 26460248 DOI: 10.1016/j.bcmd.2015.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 07/17/2015] [Accepted: 07/17/2015] [Indexed: 11/25/2022]
Abstract
Calreticulin mutation represents the second most frequent mutation after JAK2 V617F in myeloproliferative disorder and is considered to be a driving mutation. Herein the mutation burden was evaluated in patients with essential thrombocythemia or myelofibrosis and found to increase by 5.7% over time unrelated to the time elapsed from the initial to the final positive test. The longer the course of the disease when first tested (range 0-30 years, mean 7.9 years) the lower mutation burden was observed. The mutated clone was larger in type II in comparison with type I mutation when first tested but the difference in mutation burden from the final to the first positive test was significantly higher in those with type I. Similarly, the difference in mutation burden was higher in patients with essential thrombocythemia reaching almost 8% in comparison to 1.3% in post-essential thrombocythemia myelofibrosis. Thus a repeat calreticulin quantitative test is not warranted.
Collapse
Affiliation(s)
- Yulia Shuly
- Hematology Laboratory, Sheba Medical Center, Tel Hashomer, Israel
| | - Meital Nagar
- Hematology Laboratory, Sheba Medical Center, Tel Hashomer, Israel
| | - Lior Ben-Asaf
- Hematology Laboratory, Sheba Medical Center, Tel Hashomer, Israel
| | - Abraham Kneller
- Hematology Department, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - David M Steinberg
- Department of Statistics and Operations Research, Faculty of Exact Sciences, Tel Aviv University, Israel
| | | | - Ophira Salomon
- Thrombosis and Hemostasis Unit, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Israel.
| |
Collapse
|