William Vainchenker
[ INSERM UMR 1170, Gustave Roussy, Villejuif, France]
Leroy, Emilie
[UCL]
Laure Gilles
[Institut National de la Transfusion Sanguine, Paris, France]
Caroline Marty
[ INSERM UMR 1170, Gustave Roussy, Villejuif, France]
Isabelle Plo
[ INSERM UMR 1170, Gustave Roussy, Villejuif, France]
Constantinescu, Stefan N.
[UCL]
JAK inhibitors have been developed following the discovery of the JAK2V617F in 2005 as the driver mutation of the majority of non-BCR-ABL1 myeloproliferative neoplasms (MPNs). Subsequently, the search for JAK2 inhibitors continued with the discovery that the other driver mutations (CALR and MPL) also exhibited persistent JAK2 activation. Several type I ATP-competitive JAK inhibitors with different specificities were assessed in clinical trials and exhibited minimal hematologic toxicity. Interestingly, these JAK inhibitors display potent anti-inflammatory activity. Thus, JAK inhibitors targeting preferentially JAK1 and JAK3 have been developed to treat inflammation, autoimmune diseases, and graft-versus-host disease. Ten years after the beginning of clinical trials, only two drugs have been approved by the US Food and Drug Administration: one JAK2/JAK1 inhibitor (ruxolitinib) in intermediate-2 and high-risk myelofibrosis and hydroxyurea-resistant or -intolerant polycythemia vera and one JAK1/JAK3 inhibitor (tofacitinib) in methotrexate-resistant rheumatoid arthritis. The non-approved compounds exhibited many off-target effects leading to neurological and gastrointestinal toxicities, as seen in clinical trials for MPNs. Ruxolitinib is a well-tolerated drug with mostly anti-inflammatory properties. Despite a weak effect on the cause of the disease itself in MPNs, it improves the clinical state of patients and increases survival in myelofibrosis. This limited effect is related to the fact that ruxolitinib, like the other type I JAK2 inhibitors, inhibits equally mutated and wild-type JAK2 (JAK2WT) and also the JAK2 oncogenic activation. Thus, other approaches need to be developed and could be based on either (1) the development of new inhibitors specifically targeting JAK2V617F or (2) the combination of the actual JAK2 inhibitors with other therapies, in particular with molecules targeting pathways downstream of JAK2 activation or the stability of JAK2 molecule. In contrast, the strong anti-inflammatory effects of the JAK inhibitors appear as a very promising therapeutic approach for many inflammatory and auto-immune diseases.
- Quintás-Cardama A, Tong W, Kantarjian H, Thomas D, Ravandi F, Kornblau S, Manshouri T, Cortes J E, Garcia-Manero G, Verstovsek S, A phase II study of 5-azacitidine for patients with primary and post-essential thrombocythemia/polycythemia vera myelofibrosis, 10.1038/leu.2008.91
- Badar Talha, Kantarjian Hagop M., Ravandi Farhad, Jabbour Elias, Borthakur Gautam, Cortes Jorge E., Pemmaraju Naveen, Pierce Sherry R., Newberry Kate J., Daver Naval, Verstovsek Srdan, Therapeutic benefit of decitabine, a hypomethylating agent, in patients with high-risk primary myelofibrosis and myeloproliferative neoplasm in accelerated or blastic/acute myeloid leukemia phase, 10.1016/j.leukres.2015.06.001
- Dawson Mark A., Bannister Andrew J., Göttgens Berthold, Foster Samuel D., Bartke Till, Green Anthony R., Kouzarides Tony, JAK2 phosphorylates histone H3Y41 and excludes HP1α from chromatin, 10.1038/nature08448
- Liu Fan, Zhao Xinyang, Perna Fabiana, Wang Lan, Koppikar Priya, Abdel-Wahab Omar, Harr Michael W., Levine Ross L., Xu Hao, Tefferi Ayalew, Deblasio Anthony, Hatlen Megan, Menendez Silvia, Nimer Stephen D., JAK2V617F-Mediated Phosphorylation of PRMT5 Downregulates Its Methyltransferase Activity and Promotes Myeloproliferation, 10.1016/j.ccr.2010.12.020
- Schneider Rebekka K., Mullally Ann, Dugourd Aurelien, Peisker Fabian, Hoogenboezem Remco, Van Strien Paulina M.H., Bindels Eric M., Heckl Dirk, Büsche Guntram, Fleck David, Müller-Newen Gerhard, Wongboonsin Janewit, Ventura Ferreira Monica, Puelles Victor G., Saez-Rodriguez Julio, Ebert Benjamin L., Humphreys Benjamin D., Kramann Rafael, Gli1 + Mesenchymal Stromal Cells Are a Key Driver of Bone Marrow Fibrosis and an Important Cellular Therapeutic Target, 10.1016/j.stem.2017.03.008
- Sasaki Koji, Gotlib Jason R., Mesa Ruben A., Newberry Kate J., Ravandi Farhad, Cortes Jorge E., Kelly Patrick, Kutok Jeffery L., Kantarjian Hagop M., Verstovsek Srdan, Phase II evaluation of IPI-926, an oral Hedgehog inhibitor, in patients with myelofibrosis, 10.3109/10428194.2014.984703
- Stein B L, Swords R, Hochhaus A, Giles F, Novel myelofibrosis treatment strategies: potential partners for combination therapies, 10.1038/leu.2014.176
- Verstovsek Srdan, Manshouri Taghi, Pilling Darrell, Bueso-Ramos Carlos E., Newberry Kate J., Prijic Sanja, Knez Liza, Bozinovic Ksenija, Harris David M., Spaeth Erika L., Post Sean M., Multani Asha S., Rampal Raajit K., Ahn Jihae, Levine Ross L., Creighton Chad J., Kantarjian Hagop M., Estrov Zeev, Role of neoplastic monocyte-derived fibrocytes in primary myelofibrosis, 10.1084/jem.20160283
- Duffield J.S., Lupher M.R.Jr., PRM-151 (recombinant human serum amyloid P/pentraxin 2) for the treatment of fibrosis, 10.1358/dnp.2010.23.5.1444206
- Bjørn Mads Emil, Hasselbalch Hans Carl, Minimal residual disease or cure in MPNs? Rationales and perspectives on combination therapy with interferon-alpha2 and ruxolitinib, 10.1080/17474086.2017.1284583
- Hobbs Gabriela S., Hanasoge Somasundara Amritha Varshini, Kleppe Maria, Litvin Rivka, Arcila Maria, Ahn Jihae, McKenney Anna Sophia, Knapp Kristina, Ptashkin Ryan, Weinstein Howard, Heinemann Murk-Hein, Francis Jasmine, Chanel Suzanne, Berman Ellin, Mauro Michael, Tallman Martin S., Heaney Mark L., Levine Ross L., Rampal Raajit K., Hsp90 inhibition disrupts JAK-STAT signaling and leads to reductions in splenomegaly in patients with myeloproliferative neoplasms, 10.3324/haematol.2017.177600
- Mascarenhas John, Sandy Lonette, Lu Min, Yoon James, Petersen Bruce, Zhang David, Ye Fei, Newsom Carrie, Najfeld Vesna, Hochman Tsivia, Goldberg Judith D., Hoffman Ronald, A phase II study of panobinostat in patients with primary myelofibrosis (PMF) and post-polycythemia vera/essential thrombocythemia myelofibrosis (post-PV/ET MF), 10.1016/j.leukres.2016.11.015
- Marubayashi Sachie, Koppikar Priya, Taldone Tony, Abdel-Wahab Omar, West Nathan, Bhagwat Neha, Caldas-Lopes Eloisi, Ross Kenneth N., Gönen Mithat, Gozman Alex, Ahn James H., Rodina Anna, Ouerfelli Ouathek, Yang Guangbin, Hedvat Cyrus, Bradner James E., Chiosis Gabriela, Levine Ross L., HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans, 10.1172/jci42442
- Choong Meng Ling, Pecquet Christian, Pendharkar Vishal, Diaconu Carmen C., Yong Jacklyn Wei Yan, Tai Shi Jing, Wang Si Fang, Defour Jean-Philippe, Sangthongpitag Kanda, Villeval Jean-Luc, Vainchenker William, Constantinescu Stefan N., Lee May Ann, Combination treatment for myeloproliferative neoplasms using JAK and pan-class I PI3K inhibitors, 10.1111/jcmm.12156
- Bartalucci Niccolò, Tozzi Lorenzo, Bogani Costanza, Martinelli Serena, Rotunno Giada, Villeval Jean-Luc, Vannucchi Alessandro M., Co-targeting the PI3K/mTOR and JAK2 signalling pathways produces synergistic activity against myeloproliferative neoplasms, 10.1111/jcmm.12162
- Lai Ping-Shan, Rosa David A, Magdy Ali Ahmed, Gómez-Biagi Rodolfo F, Ball Daniel P, Shouksmith Andrew E, Gunning Patrick T, A STAT inhibitor patent review: progress since 2011, 10.1517/13543776.2015.1086749
- Ihle J N, Witthuhn B A, Quelle F W, Yamamoto K, Silvennoinen O, Signaling Through the Hematopoietic Cytokine Receptors, 10.1146/annurev.iy.13.040195.002101
- McMullin Mary Frances, Harrison Claire N., Niederwieser Dietger, Demuynck Hilde, Jäkel Nadja, Gopalakrishna Prashanth, McQuitty Mari, Stalbovskaya Viktoriya, Recher Christian, Theunissen Koen, Gisslinger Heinz, Kiladjian Jean-Jacques, Al-Ali Haifa-Kathrin, The use of erythropoiesis-stimulating agents with ruxolitinib in patients with myelofibrosis in COMFORT-II: an open-label, phase 3 study assessing efficacy and safety of ruxolitinib versus best available therapy in the treatment of myelofibrosis, 10.1186/s40164-015-0021-2
- Dussiot Michael, Maciel Thiago T, Fricot Aurélie, Chartier Céline, Negre Olivier, Veiga Joel, Grapton Damien, Paubelle Etienne, Payen Emmanuel, Beuzard Yves, Leboulch Philippe, Ribeil Jean-Antoine, Arlet Jean-Benoit, Coté Francine, Courtois Geneviève, Ginzburg Yelena Z, Daniel Thomas O, Chopra Rajesh, Sung Victoria, Hermine Olivier, Moura Ivan C, An activin receptor IIA ligand trap corrects ineffective erythropoiesis in β-thalassemia, 10.1038/nm.3468
- Thomas Sally, Fisher Katherine H., Snowden John A., Danson Sarah J., Brown Stephen, Zeidler Martin P., Methotrexate Is a JAK/STAT Pathway Inhibitor, 10.1371/journal.pone.0130078
- Dusa Alexandra, Staerk Judith, Elliott Joanne, Pecquet Christian, Poirel Hélène A., Johnston James A., Constantinescu Stefan N., Substitution of Pseudokinase Domain Residue Val-617 by Large Non-polar Amino Acids Causes Activation of JAK2, 10.1074/jbc.m709302200
- Puleo David E., Kucera Kaury, Hammarén Henrik M., Ungureanu Daniela, Newton Ana S., Silvennoinen Olli, Jorgensen William L., Schlessinger Joseph, Identification and Characterization of JAK2 Pseudokinase Domain Small Molecule Binders, 10.1021/acsmedchemlett.7b00153
- Bandaranayake Rajintha M, Ungureanu Daniela, Shan Yibing, Shaw David E, Silvennoinen Olli, Hubbard Stevan R, Crystal structures of the JAK2 pseudokinase domain and the pathogenic mutant V617F, 10.1038/nsmb.2348
- Ohren Jeffrey F, Chen Huifen, Pavlovsky Alexander, Whitehead Christopher, Zhang Erli, Kuffa Peter, Yan Chunhong, McConnell Patrick, Spessard Cindy, Banotai Craig, Mueller W Thomas, Delaney Amy, Omer Charles, Sebolt-Leopold Judith, Dudley David T, Leung Iris K, Flamme Cathlin, Warmus Joseph, Kaufman Michael, Barrett Stephen, Tecle Haile, Hasemann Charles A, Structures of human MAP kinase kinase 1 (MEK1) and MEK2 describe novel noncompetitive kinase inhibition, 10.1038/nsmb859
- Zhang Jianming, Adrián Francisco J., Jahnke Wolfgang, Cowan-Jacob Sandra W., Li Allen G., Iacob Roxana E., Sim Taebo, Powers John, Dierks Christine, Sun Fangxian, Guo Gui-Rong, Ding Qiang, Okram Barun, Choi Yongmun, Wojciechowski Amy, Deng Xianming, Liu Guoxun, Fendrich Gabriele, Strauss André, Vajpai Navratna, Grzesiek Stephan, Tuntland Tove, Liu Yi, Bursulaya Badry, Azam Mohammad, Manley Paul W., Engen John R., Daley George Q., Warmuth Markus, Gray Nathanael S., Targeting Bcr–Abl by combining allosteric with ATP-binding-site inhibitors, 10.1038/nature08675
- Hornakova T., Springuel L., Devreux J., Dusa A., Constantinescu S. N., Knoops L., Renauld J.-C., Oncogenic JAK1 and JAK2-activating mutations resistant to ATP-competitive inhibitors, 10.3324/haematol.2010.036350
- Deshpande A, Reddy M M, Schade G O M, Ray A, Chowdary T K, Griffin J D, Sattler M, Kinase domain mutations confer resistance to novel inhibitors targeting JAK2V617F in myeloproliferative neoplasms, 10.1038/leu.2011.255
- Haan Claude, Rolvering Catherine, Raulf Friedrich, Kapp Manuela, Drückes Peter, Thoma Gebhard, Behrmann Iris, Zerwes Hans-Günter, Jak1 Has a Dominant Role over Jak3 in Signal Transduction through γc-Containing Cytokine Receptors, 10.1016/j.chembiol.2011.01.012
- Genovese Mark C, Yang Fang, Østergaard Mikkel, Kinnman Nils, Efficacy of VX-509 (decernotinib) in combination with a disease-modifying antirheumatic drug in patients with rheumatoid arthritis: clinical and MRI findings, 10.1136/annrheumdis-2015-208901
- Richez Christophe, Truchetet Marie-Elise, Kostine Marie, Schaeverbeke Thierry, Bannwarth Bernard, Efficacy of baricitinib in the treatment of rheumatoid arthritis, 10.1080/14656566.2017.1359256
- Spoerl S., Mathew N. R., Bscheider M., Schmitt-Graeff A., Chen S., Mueller T., Verbeek M., Fischer J., Otten V., Schmickl M., Maas-Bauer K., Finke J., Peschel C., Duyster J., Poeck H., Zeiser R., von Bubnoff N., Activity of therapeutic JAK 1/2 blockade in graft-versus-host disease, 10.1182/blood-2013-12-543736
- Costa Luisa, Del Puente Antonio, Peluso Rosario, Tasso Marco, Caso Paolo, Chimenti Maria Sole, Sabbatino Vincenzo, Girolimetto Nicolò, Benigno Carolina, Bertolini Nicoletta, Del Puente Aurora, Perricone Roberto, Scarpa Raffaele, Caso Francesco, Small molecule therapy for managing moderate to severe psoriatic arthritis, 10.1080/14656566.2017.1378343
- Archer Thomas P, Moran Gordon W, Ghosh Subrata, Tofacitinib in ulcerative colitis, 10.2217/imt-2015-0031
- Winthrop Kevin L., The emerging safety profile of JAK inhibitors in rheumatic disease, 10.1038/nrrheum.2017.23
- Welsch Katharina, Holstein Julia, Laurence Arian, Ghoreschi Kamran, Targeting JAK/STAT signalling in inflammatory skin diseases with small molecule inhibitors, 10.1002/eji.201646680
- Schwaab Juliana, Knut Marcin, Haferlach Claudia, Metzgeroth Georgia, Horny Hans-Peter, Chase Andrew, Tapper William, Score Joannah, Waghorn Katherine, Naumann Nicole, Jawhar Mohamad, Fabarius Alice, Hofmann Wolf-Karsten, Cross Nicholas C. P., Reiter Andreas, Limited duration of complete remission on ruxolitinib in myeloid neoplasms with PCM1-JAK2 and BCR-JAK2 fusion genes, 10.1007/s00277-014-2221-y
- Rumi Elisa, Milosevic Jelena D., Selleslag Dominik, Casetti Ilaria, Lierman Els, Pietra Daniela, Cavalloni Chiara, Bellini Marta, Milanesi Chiara, Dambruoso Irene, Astori Cesare, Kralovics Robert, Vandenberghe Peter, Cazzola Mario, Efficacy of ruxolitinib in myeloid neoplasms with PCM1-JAK2 fusion gene, 10.1007/s00277-015-2451-7
- Lierman E., Selleslag D., Smits S., Billiet J., Vandenberghe P., Ruxolitinib inhibits transforming JAK2 fusion proteins in vitro and induces complete cytogenetic remission in t(8;9)(p22;p24)/PCM1-JAK2-positive chronic eosinophilic leukemia, 10.1182/blood-2012-06-433821
- Dao Kim-Hien T., Solti Magdolna B., Maxson Julia E., Winton Elliott F., Press Richard D., Druker Brian J., Tyner Jeffrey W., Significant clinical response to JAK1/2 inhibition in a patient with CSF3R-T618I-positive atypical chronic myeloid leukemia, 10.1016/j.lrr.2014.07.002
- Yu Helena A., Perez Leslie, Chang Qing, Gao Sizhi P., Kris Mark G., Riely Gregory J., Bromberg Jacqueline, A Phase 1/2 Trial of Ruxolitinib and Erlotinib in Patients with EGFR -Mutant Lung Adenocarcinomas with Acquired Resistance to Erlotinib, 10.1016/j.jtho.2016.08.140
- Buchert M, Burns C J, Ernst M, Targeting JAK kinase in solid tumors: emerging opportunities and challenges, 10.1038/onc.2015.150
- Hazell Alan S., Afadlal Szeifoul, Cheresh David A., Azar Ashraf, Treatment of rats with the JAK-2 inhibitor fedratinib does not lead to experimental Wernicke’s encephalopathy, 10.1016/j.neulet.2017.01.041
- Pardanani Animesh, Harrison Claire, Cortes Jorge E., Cervantes Francisco, Mesa Ruben A., Milligan Donald, Masszi Tamás, Mishchenko Elena, Jourdan Eric, Vannucchi Alessandro M., Drummond Mark W., Jurgutis Mindaugas, Kuliczkowski Kazimierz, Gheorghita Emanuil, Passamonti Francesco, Neumann Frank, Patki Abhay, Gao Guozhi, Tefferi Ayalew, Safety and Efficacy of Fedratinib in Patients With Primary or Secondary Myelofibrosis : A Randomized Clinical Trial, 10.1001/jamaoncol.2015.1590
- Jamieson Catriona, Hasserjian Robert, Gotlib Jason, Cortes Jorge, Stone Richard, Talpaz Moshe, Thiele Jürgen, Rodig Scott, Pozdnyakova Olga, Effect of treatment with a JAK2-selective inhibitor, fedratinib, on bone marrow fibrosis in patients with myelofibrosis, 10.1186/s12967-015-0644-4
- Harrison Claire N, Schaap Nicolaas, Vannucchi Alessandro M, Kiladjian Jean-Jacques, Tiu Ramon V, Zachee Pierre, Jourdan Eric, Winton Elliott, Silver Richard T, Schouten Harry C, Passamonti Francesco, Zweegman Sonja, Talpaz Moshe, Lager Joanne, Shun Zhenming, Mesa Ruben A, Janus kinase-2 inhibitor fedratinib in patients with myelofibrosis previously treated with ruxolitinib (JAKARTA-2): a single-arm, open-label, non-randomised, phase 2, multicentre study, 10.1016/s2352-3026(17)30088-1
- (2017)
- (2016)
- Verstovsek Srdan, Odenike Olatoyosi, Singer Jack W., Granston Tanya, Al-Fayoumi Suliman, Deeg H. Joachim, Phase 1/2 study of pacritinib, a next generation JAK2/FLT3 inhibitor, in myelofibrosis or other myeloid malignancies, 10.1186/s13045-016-0367-x
- Verstovsek S, Talpaz M, Ritchie E, Wadleigh M, Odenike O, Jamieson C, Stein B, Uno T, Mesa R A, A phase I, open-label, dose-escalation, multicenter study of the JAK2 inhibitor NS-018 in patients with myelofibrosis, 10.1038/leu.2016.215
- Asshoff Malte, Petzer Verena, Warr Matthew R., Haschka David, Tymoszuk Piotr, Demetz Egon, Seifert Markus, Posch Wilfried, Nairz Manfred, Maciejewski Pat, Fowles Peter, Burns Christopher J., Smith Gregg, Wagner Kay-Uwe, Weiss Guenter, Whitney J. Andrew, Theurl Igor, Momelotinib inhibits ACVR1/ALK2, decreases hepcidin production, and ameliorates anemia of chronic disease in rodents, 10.1182/blood-2016-09-740092
- A Pardanani, Blood., 122, 108 (2013)
- Gupta Vikas, Mesa Ruben A., Deininger Michael W.N., Rivera Candido E., Sirhan Shireen, Brachmann Carrie Baker, Collins Helen, Kawashima Jun, Xin Yan, Verstovsek Srdan, A phase 1/2, open-label study evaluating twice-daily administration of momelotinib in myelofibrosis, 10.3324/haematol.2016.148924
- Pardanani A, Abdelrahman R A, Finke C, Lasho T T, Begna K H, Al-Kali A, Hogan W J, Litzow M R, Hanson C A, Ketterling R P, Tefferi A, Genetic determinants of response and survival in momelotinib-treated patients with myelofibrosis, 10.1038/leu.2014.306
- Verstovsek Srdan, Gotlib Jason, Mesa Ruben A., Vannucchi Alessandro M., Kiladjian Jean-Jacques, Cervantes Francisco, Harrison Claire N., Paquette Ronald, Sun William, Naim Ahmad, Langmuir Peter, Dong Tuochuan, Gopalakrishna Prashanth, Gupta Vikas, Long-term survival in patients treated with ruxolitinib for myelofibrosis: COMFORT-I and -II pooled analyses, 10.1186/s13045-017-0527-7
- Curran Shane A., Shyer Justin A., St. Angelo Erin T., Talbot Lillian R., Sharma Sneh, Chung David J., Heller Glenn, Hsu Katharine C., Betts Brian C., Young James W., Human Dendritic Cells Mitigate NK-Cell Dysfunction Mediated by Nonselective JAK1/2 Blockade, 10.1158/2326-6066.cir-16-0233
- Schonberg K., Rudolph J., Vonnahme M., Parampalli Yajnanarayana S., Cornez I., Hejazi M., Manser A. R., Uhrberg M., Verbeek W., Koschmieder S., Brummendorf T. H., Brossart P., Heine A., Wolf D., JAK Inhibition Impairs NK Cell Function in Myeloproliferative Neoplasms, 10.1158/0008-5472.can-14-3198
- Saeed Iram, McLornan Donal, Harrison Claire N., Managing side effects of JAK inhibitors for myelofibrosis in clinical practice, 10.1080/17474086.2017.1337507
- Curto-Garcia Natalia, Harrison Claire N, An updated review of the JAK1/2 inhibitor (ruxolitinib) in the Philadelphia-negative myeloproliferative neoplasms, 10.2217/fon-2017-0298
- Verstovsek Srdan, Passamonti Francesco, Rambaldi Alessandro, Barosi Giovanni, Rumi Elisa, Gattoni Elisabetta, Pieri Lisa, Zhen Huiling, Granier Muriel, Assad Albert, Cazzola Mario, Kantarjian Hagop M., Barbui Tiziano, Vannucchi Alessandro M., Ruxolitinib for essential thrombocythemia refractory to or intolerant of hydroxyurea: long-term phase 2 study results, 10.1182/blood-2017-02-765032
- Harrison Claire N., Mead Adam J., Panchal Anesh, Fox Sonia, Yap Christina, Gbandi Emmanouela, Houlton Aimee, Alimam Samah, Ewing Joanne, Wood Marion, Chen Frederick, Coppell Jason, Panoskaltsis Nicki, Knapper Steven, Ali Sahra, Hamblin Angela, Scherber Robyn, Dueck Amylou C., Cross Nicholas C. P., Mesa Ruben, McMullin Mary Frances, Ruxolitinib vs best available therapy for ET intolerant or resistant to hydroxycarbamide, 10.1182/blood-2017-05-785790
- Passamonti Francesco, Griesshammer Martin, Palandri Francesca, Egyed Miklos, Benevolo Giulia, Devos Timothy, Callum Jeannie, Vannucchi Alessandro M, Sivgin Serdar, Bensasson Caroline, Khan Mahmudul, Mounedji Nadjat, Saydam Guray, Ruxolitinib for the treatment of inadequately controlled polycythaemia vera without splenomegaly (RESPONSE-2): a randomised, open-label, phase 3b study, 10.1016/s1470-2045(16)30558-7
- Al-Ali H. K., Griesshammer M., le Coutre P., Waller C. F., Liberati A. M., Schafhausen P., Tavares R., Giraldo P., Foltz L., Raanani P., Gupta V., Tannir B., Ronco J. P., Ghosh J., Martino B., Vannucchi A. M., Safety and efficacy of ruxolitinib in an open-label, multicenter, single-arm phase 3b expanded-access study in patients with myelofibrosis: a snapshot of 1144 patients in the JUMP trial, 10.3324/haematol.2016.143677
- Jatiani S. S., Cosenza S. C., Reddy M. V. R., Ha J. H., Baker S. J., Samanta A. K., Olnes M. J., Pfannes L., Sloand E. M., Arlinghaus R. B., Reddy E. P., A Non-ATP-Competitive Dual Inhibitor of JAK2V617F and BCR-ABLT315I Kinases: Elucidation of a Novel Therapeutic Spectrum Based on Substrate Competitive Inhibition, 10.1177/1947601910371337
- Lipka D. B., Hoffmann L. S., Heidel F., Markova B., Blum M.-C., Breitenbuecher F., Kasper S., Kindler T., Levine R. L., Huber C., Fischer T., LS104, a non-ATP-competitive small-molecule inhibitor of JAK2, is potently inducing apoptosis in JAK2V617F-positive cells, 10.1158/1535-7163.mct-07-2215
- Takeuchi Tsutomu, Tanaka Yoshiya, Iwasaki Manabu, Ishikura Hiroaki, Saeki Satoshi, Kaneko Yuichiro, Efficacy and safety of the oral Janus kinase inhibitor peficitinib (ASP015K) monotherapy in patients with moderate to severe rheumatoid arthritis in Japan: a 12-week, randomised, double-blind, placebo-controlled phase IIb study, 10.1136/annrheumdis-2015-208279
- Papp K., Pariser D., Catlin M., Wierz G., Ball G., Akinlade B., Zeiher B., Krueger J.G., A phase 2a randomized, double-blind, placebo-controlled, sequential dose-escalation study to evaluate the efficacy and safety of ASP015K, a novel Janus kinase inhibitor, in patients with moderate-to-severe psoriasis, 10.1111/bjd.13745
- Uckun F. M., Janus kinase 3 inhibitor WHI-P131/JANEX-1 prevents graft-versus-host disease but spares the graft-versus-leukemia function of the bone marrow allografts in a murine bone marrow transplantation model, 10.1182/blood.v99.11.4192
- Fleischmann Roy M., Damjanov Nemanja S., Kivitz Alan J., Legedza Anna, Hoock Thomas, Kinnman Nils, A Randomized, Double-Blind, Placebo-Controlled, Twelve-Week, Dose-Ranging Study of Decernotinib, an Oral Selective JAK-3 Inhibitor, as Monotherapy in Patients With Active Rheumatoid Arthritis : Decernotinib as Monotherapy in Patients With RA, 10.1002/art.38949
- Klünder Ben, Mohamed Mohamed-Eslam F., Othman Ahmed A., Population Pharmacokinetics of Upadacitinib in Healthy Subjects and Subjects with Rheumatoid Arthritis: Analyses of Phase I and II Clinical Trials, 10.1007/s40262-017-0605-6
- Schmieder G.J., Draelos Z.D., Pariser D.M., Banfield C., Cox L., Hodge M., Kieras E., Parsons-Rich D., Menon S., Salganik M., Page K., Peeva E., Efficacy and safety of the Janus Kinase 1 inhibitor PF-04965842 in patients with moderate to severe psoriasis: phase 2, randomized, double-blind, placebo-controlled study, 10.1111/bjd.16004
- Bissonnette Robert, Luchi Monica, Fidelus-Gort Rosanne, Jackson Shawnta, Zhang Haifeng, Flores Robert, Newton Robert, Scherle Peggy, Yeleswaram Swamy, Chen Xuejun, Menter Alan, A randomized, double-blind, placebo-controlled, dose-escalation study of the safety and efficacy of INCB039110, an oral janus kinase 1 inhibitor, in patients with stable, chronic plaque psoriasis, 10.3109/09546634.2015.1115819
- Mascarenhas John O., Talpaz Moshe, Gupta Vikas, Foltz Lynda M., Savona Michael R., Paquette Ronald, Turner A. Robert, Coughlin Paul, Winton Elliott, Burn Timothy C., O’Neill Peter, Clark Jason, Hunter Deborah, Assad Albert, Hoffman Ronald, Verstovsek Srdan, Primary analysis of a phase II open-label trial of INCB039110, a selective JAK1 inhibitor, in patients with myelofibrosis, 10.3324/haematol.2016.151126
- Baker Kenneth F, Isaacs John D, Novel therapies for immune-mediated inflammatory diseases: What can we learn from their use in rheumatoid arthritis, spondyloarthritis, systemic lupus erythematosus, psoriasis, Crohn’s disease and ulcerative colitis?, 10.1136/annrheumdis-2017-211555
- Vermeire Séverine, Schreiber Stefan, Petryka Robert, Kuehbacher Tanja, Hebuterne Xavier, Roblin Xavier, Klopocka Maria, Goldis Adrian, Wisniewska-Jarosinska Maria, Baranovsky Andrey, Sike Robert, Stoyanova Kremena, Tasset Chantal, Van der Aa Annegret, Harrison Pille, Clinical remission in patients with moderate-to-severe Crohn's disease treated with filgotinib (the FITZROY study): results from a phase 2, double-blind, randomised, placebo-controlled trial, 10.1016/s0140-6736(16)32537-5
- Vanhoutte Frédéric, Mazur Minodora, Voloshyn Oleksandr, Stanislavchuk Mykola, Van der Aa Annegret, Namour Florence, Galien René, Meuleners Luc, van 't Klooster Gerben, Efficacy, Safety, Pharmacokinetics, and Pharmacodynamics of Filgotinib, a Selective JAK-1 Inhibitor, After Short-Term Treatment of Rheumatoid Arthritis: Results of Two Randomized Phase IIa Trials : CLINICAL EXPLORATION OF FILGOTINIB IN RA, 10.1002/art.40186
- Pardanani A, Tefferi A, Jamieson C, Gabrail N Y, Lebedinsky C, Gao G, Liu F, Xu C, Cao H, Talpaz M, A phase 2 randomized dose-ranging study of the JAK2-selective inhibitor fedratinib (SAR302503) in patients with myelofibrosis, 10.1038/bcj.2015.63
- Pardanani A, Hood J, Lasho T, Levine R L, Martin M B, Noronha G, Finke C, Mak C C, Mesa R, Zhu H, Soll R, Gilliland D G, Tefferi A, TG101209, a small molecule JAK2-selective kinase inhibitor potently inhibits myeloproliferative disorder-associated JAK2V617F and MPLW515L/K mutations, 10.1038/sj.leu.2404750
- Ringel Frauke, Kaeda Jaspal, Schwarz Michaela, Oberender Christian, Grille Peggy, Dörken Bernd, Marque Fanny, Manley Paul W., Radimerski Thomas, le Coutre Philipp, Effects of Jak2 Type 1 Inhibitors NVP-BSK805 and NVP-BVB808 on Jak2 Mutation-Positive and Bcr-Abl-Positive Cell Lines, 10.1159/000356784
- Stump Kristine L, Lu Lily D, Dobrzanski Pawel, Serdikoff Cynthia, Gingrich Diane E, Dugan Ben J, Angeles Thelma S, Albom Mark S, Ator Mark A, Dorsey Bruce D, Ruggeri Bruce A, Seavey Matthew M, A highly selective, orally active inhibitor of Janus kinase 2, CEP-33779, ablates disease in two mouse models of rheumatoid arthritis, 10.1186/ar3329
- J Mascarenhas (2016)
- Mesa Ruben A, Vannucchi Alessandro M, Mead Adam, Egyed Miklos, Szoke Anita, Suvorov Aleksandr, Jakucs Janos, Perkins Andrew, Prasad Ritam, Mayer Jiri, Demeter Judit, Ganly Peter, Singer Jack W, Zhou Huafeng, Dean James P, te Boekhorst Peter A, Nangalia Jyoti, Kiladjian Jean-Jacques, Harrison Claire N, Pacritinib versus best available therapy for the treatment of myelofibrosis irrespective of baseline cytopenias (PERSIST-1): an international, randomised, phase 3 trial, 10.1016/s2352-3026(17)30027-3
- Nakaya Y, Shide K, Niwa T, Homan J, Sugahara S, Horio T, Kuramoto K, Kotera T, Shibayama H, Hori K, Naito H, Shimoda K, Efficacy of NS-018, a potent and selective JAK2/Src inhibitor, in primary cells and mouse models of myeloproliferative neoplasms, 10.1038/bcj.2011.29
- Baffert F., Regnier C. H., De Pover A., Pissot-Soldermann C., Tavares G. A., Blasco F., Brueggen J., Chene P., Drueckes P., Erdmann D., Furet P., Gerspacher M., Lang M., Ledieu D., Nolan L., Ruetz S., Trappe J., Vangrevelinghe E., Wartmann M., Wyder L., Hofmann F., Radimerski T., Potent and Selective Inhibition of Polycythemia by the Quinoxaline JAK2 Inhibitor NVP-BSK805, 10.1158/1535-7163.mct-10-0053
- Verstovsek Srdan, Tam Constantine S., Wadleigh Martha, Sokol Lubomir, Smith Catherine C., Bui Lynne A., Song Chunyan, Clary Douglas O., Olszynski Patrycja, Cortes Jorge, Kantarjian Hagop, Shah Neil P., Phase I evaluation of XL019, an oral, potent, and selective JAK2 inhibitor, 10.1016/j.leukres.2013.12.006
- Verstovsek Srdan, Mesa Ruben A., Salama Mohamed E., Li Li, Pitou Celine, Nunes Fabio P., Price Gregory L., Giles Jennifer L., D’Souza Deborah N., Walgren Richard A., Prchal Josef T., A phase 1 study of the Janus kinase 2 ( JAK2 ) V617F inhibitor, gandotinib (LY2784544), in patients with primary myelofibrosis, polycythemia vera, and essential thrombocythemia, 10.1016/j.leukres.2017.08.010
- Vu M., Heyes C., Robertson S. J., Varigos G. A., Ross G., Oral tofacitinib: a promising treatment in atopic dermatitis, alopecia areata and vitiligo, 10.1111/ced.13290
- Feldman Steven R., Thaçi Diamant, Gooderham Melinda, Augustin Matthias, de la Cruz Claudia, Mallbris Lotus, Buonanno Marjorie, Tatulych Svitlana, Kaur Mandeep, Lan Shuping, Valdez Hernan, Mamolo Carla, Tofacitinib improves pruritus and health-related quality of life up to 52 weeks: Results from 2 randomized phase III trials in patients with moderate to severe plaque psoriasis, 10.1016/j.jaad.2016.07.040
- Panés Julian, Sandborn William J, Schreiber Stefan, Sands Bruce E, Vermeire Séverine, D'Haens Geert, Panaccione Remo, Higgins Peter D R, Colombel Jean-Frederic, Feagan Brian G, Chan Gary, Moscariello Michele, Wang Wenjin, Niezychowski Wojciech, Marren Amy, Healey Paul, Maller Eric, Tofacitinib for induction and maintenance therapy of Crohn's disease: results of two phase IIb randomised placebo-controlled trials, 10.1136/gutjnl-2016-312735
- NICE issues positive recommendation for XELJANZ®▼(tofacitinib citrate) as a new treatment option for adults with severe rheumatoid arthritis, 10.1093/rheumatology/kex407
- Papp K.A., Menter M.A., Raman M., Disch D., Schlichting D.E., Gaich C., Macias W., Zhang X., Janes J.M., A randomized phase 2b trial of baricitinib, an oral Janus kinase (JAK) 1/JAK2 inhibitor, in patients with moderate-to-severe psoriasis, 10.1111/bjd.14403
- Keystone Edward C, Taylor Peter C, Tanaka Yoshiya, Gaich Carol, DeLozier Amy M, Dudek Anna, Zamora Jorge Velasco, Cobos Jose Arturo Covarrubias, Rooney Terence, Bono Stephanie de, Arora Vipin, Linetzky Bruno, Weinblatt Michael E, Patient-reported outcomes from a phase 3 study of baricitinib versus placebo or adalimumab in rheumatoid arthritis: secondary analyses from the RA-BEAM study, 10.1136/annrheumdis-2017-211259
- Plimack E. R., LoRusso P. M., McCoon P., Tang W., Krebs A. D., Curt G., Eckhardt S. G., AZD1480: A Phase I Study of a Novel JAK2 Inhibitor in Solid Tumors, 10.1634/theoncologist.2013-0198
- Suryani S., Bracken L. S., Harvey R. C., Sia K. C. S., Carol H., Chen I.-M., Evans K., Dietrich P. A., Roberts K. G., Kurmasheva R. T., Billups C. A., Mullighan C. G., Willman C. L., Loh M. L., Hunger S. P., Houghton P. J., Smith M. A., Lock R. B., Evaluation of the In Vitro and In Vivo Efficacy of the JAK Inhibitor AZD1480 against JAK-Mutated Acute Lymphoblastic Leukemia, 10.1158/1535-7163.mct-14-0647
- Verstovsek Srdan, Hoffman Ronald, Mascarenhas John, Soria Jean-Charles, Bahleda Ratislav, McCoon Patricia, Tang Weifeng, Cortes Jorge, Kantarjian Hagop, Ribrag Vincent, A phase I, open-label, multi-center study of the JAK2 inhibitor AZD1480 in patients with myelofibrosis, 10.1016/j.leukres.2014.11.018
- Verstovsek Srdan, Courby Stephane, Griesshammer Martin, Mesa Ruben A., Brachmann Carrie Baker, Kawashima Jun, Maltzman Julia D., Shao Lixin, Xin Yan, Huang Daniel, Bajel Ashish, A phase 2 study of momelotinib, a potent JAK1 and JAK2 inhibitor, in patients with polycythemia vera or essential thrombocythemia, 10.1016/j.leukres.2017.05.002
- Mesa Ruben A., Kiladjian Jean-Jacques, Catalano John V., Devos Timothy, Egyed Miklos, Hellmann Andrzei, McLornan Donal, Shimoda Kazuya, Winton Elliott F., Deng Wei, Dubowy Ronald L., Maltzman Julia D., Cervantes Francisco, Gotlib Jason, SIMPLIFY-1: A Phase III Randomized Trial of Momelotinib Versus Ruxolitinib in Janus Kinase Inhibitor–Naïve Patients With Myelofibrosis, 10.1200/jco.2017.73.4418
- Harris John E., Rashighi Mehdi, Nguyen Nhan, Jabbari Ali, Ulerio Grace, Clynes Raphael, Christiano Angela M., Mackay-Wiggan Julian, Rapid skin repigmentation on oral ruxolitinib in a patient with coexistent vitiligo and alopecia areata (AA), 10.1016/j.jaad.2015.09.073
- Mackay-Wiggan Julian, Jabbari Ali, Nguyen Nhan, Cerise Jane E., Clark Charlotte, Ulerio Grace, Furniss Megan, Vaughan Roger, Christiano Angela M., Clynes Raphael, Oral ruxolitinib induces hair regrowth in patients with moderate-to-severe alopecia areata, 10.1172/jci.insight.89790
- Punwani Naresh, Scherle Peggy, Flores Robert, Shi Jack, Liang Jinjin, Yeleswaram Swamy, Levy Richard, Williams William, Gottlieb Alice, Preliminary clinical activity of a topical JAK1/2 inhibitor in the treatment of psoriasis, 10.1016/j.jaad.2011.12.018
- Zeiser R, Burchert A, Lengerke C, Verbeek M, Maas-Bauer K, Metzelder S K, Spoerl S, Ditschkowski M, Ecsedi M, Sockel K, Ayuk F, Ajib S, de Fontbrune F S, Na I-K, Penter L, Holtick U, Wolf D, Schuler E, Meyer E, Apostolova P, Bertz H, Marks R, Lübbert M, Wäsch R, Scheid C, Stölzel F, Ordemann R, Bug G, Kobbe G, Negrin R, Brune M, Spyridonidis A, Schmitt-Gräff A, van der Velden W, Huls G, Mielke S, Grigoleit G U, Kuball J, Flynn R, Ihorst G, Du J, Blazar B R, Arnold R, Kröger N, Passweg J, Halter J, Socié G, Beelen D, Peschel C, Neubauer A, Finke J, Duyster J, von Bubnoff N, Ruxolitinib in corticosteroid-refractory graft-versus-host disease after allogeneic stem cell transplantation: a multicenter survey, 10.1038/leu.2015.212
- Hurwitz Herbert I., Uppal Nikhil, Wagner Stephanie A., Bendell Johanna C., Beck J. Thaddeus, Wade Seaborn M., Nemunaitis John J., Stella Philip J., Pipas J. Marc, Wainberg Zev A., Manges Robert, Garrett William M., Hunter Deborah S., Clark Jason, Leopold Lance, Sandor Victor, Levy Richard S., Randomized, Double-Blind, Phase II Study of Ruxolitinib or Placebo in Combination With Capecitabine in Patients With Metastatic Pancreatic Cancer for Whom Therapy With Gemcitabine Has Failed, 10.1200/jco.2015.61.4578
- Eghtedar A., Verstovsek S., Estrov Z., Burger J., Cortes J., Bivins C., Faderl S., Ferrajoli A., Borthakur G., George S., Scherle P. A., Newton R. C., Kantarjian H. M., Ravandi F., Phase 2 study of the JAK kinase inhibitor ruxolitinib in patients with refractory leukemias, including postmyeloproliferative neoplasm acute myeloid leukemia, 10.1182/blood-2011-12-400051
- Verstovsek Srdan, Mesa Ruben A., Gotlib Jason, Levy Richard S., Gupta Vikas, DiPersio John F., Catalano John V., Deininger Michael, Miller Carole, Silver Richard T., Talpaz Moshe, Winton Elliott F., Harvey Jimmie H., Arcasoy Murat O., Hexner Elizabeth, Lyons Roger M., Paquette Ronald, Raza Azra, Vaddi Kris, Erickson-Viitanen Susan, Koumenis Iphigenia L., Sun William, Sandor Victor, Kantarjian Hagop M., A Double-Blind, Placebo-Controlled Trial of Ruxolitinib for Myelofibrosis, 10.1056/nejmoa1110557
- Vannucchi Alessandro M., Kiladjian Jean Jacques, Griesshammer Martin, Masszi Tamas, Durrant Simon, Passamonti Francesco, Harrison Claire N., Pane Fabrizio, Zachee Pierre, Mesa Ruben, He Shui, Jones Mark M., Garrett William, Li Jingjin, Pirron Ulrich, Habr Dany, Verstovsek Srdan, Ruxolitinib versus Standard Therapy for the Treatment of Polycythemia Vera, 10.1056/nejmoa1409002
- Harrison Claire, Kiladjian Jean-Jacques, Al-Ali Haifa Kathrin, Gisslinger Heinz, Waltzman Roger, Stalbovskaya Viktoriya, McQuitty Mari, Hunter Deborah S., Levy Richard, Knoops Laurent, Cervantes Francisco, Vannucchi Alessandro M., Barbui Tiziano, Barosi Giovanni, JAK Inhibition with Ruxolitinib versus Best Available Therapy for Myelofibrosis, 10.1056/nejmoa1110556
- Wu Shuo-Chieh, Li Loretta S., Kopp Nadja, Montero Joan, Chapuy Bjoern, Yoda Akinori, Christie Amanda L., Liu Huiyun, Christodoulou Alexandra, van Bodegom Diederik, van der Zwet Jordy, Layer Jacob V., Tivey Trevor, Lane Andrew A., Ryan Jeremy A., Ng Samuel Y., DeAngelo Daniel J., Stone Richard M., Steensma David, Wadleigh Martha, Harris Marian, Mandon Emeline, Ebel Nicolas, Andraos Rita, Romanet Vincent, Dölemeyer Arno, Sterker Dario, Zender Michael, Rodig Scott J., Murakami Masato, Hofmann Francesco, Kuo Frank, Eck Michael J., Silverman Lewis B., Sallan Stephen E., Letai Anthony, Baffert Fabienne, Vangrevelinghe Eric, Radimerski Thomas, Gaul Christoph, Weinstock David M., Activity of the Type II JAK2 Inhibitor CHZ868 in B Cell Acute Lymphoblastic Leukemia, 10.1016/j.ccell.2015.06.005
- Meyer Sara C., Keller Matthew D., Chiu Sophia, Koppikar Priya, Guryanova Olga A., Rapaport Franck, Xu Ke, Manova Katia, Pankov Dmitry, O’Reilly Richard J., Kleppe Maria, McKenney Anna Sophia, Shih Alan H., Shank Kaitlyn, Ahn Jihae, Papalexi Eftymia, Spitzer Barbara, Socci Nick, Viale Agnes, Mandon Emeline, Ebel Nicolas, Andraos Rita, Rubert Joëlle, Dammassa Ernesta, Romanet Vincent, Dölemeyer Arno, Zender Michael, Heinlein Melanie, Rampal Raajit, Weinberg Rona Singer, Hoffman Ronald, Sellers William R., Hofmann Francesco, Murakami Masato, Baffert Fabienne, Gaul Christoph, Radimerski Thomas, Levine Ross L., CHZ868, a Type II JAK2 Inhibitor, Reverses Type I JAK Inhibitor Persistence and Demonstrates Efficacy in Myeloproliferative Neoplasms, 10.1016/j.ccell.2015.06.006
- Blanc Javier, Geney Raphael, Menet Christel, Type II Kinase Inhibitors: An Opportunity in Cancer for Rational Design, 10.2174/1871520611313050008
- Meyer S. C., Levine R. L., Molecular Pathways: Molecular Basis for Sensitivity and Resistance to JAK Kinase Inhibitors, 10.1158/1078-0432.ccr-13-0279
- Koppikar Priya, Bhagwat Neha, Kilpivaara Outi, Manshouri Taghi, Adli Mazhar, Hricik Todd, Liu Fan, Saunders Lindsay M., Mullally Ann, Abdel-Wahab Omar, Leung Laura, Weinstein Abby, Marubayashi Sachie, Goel Aviva, Gönen Mithat, Estrov Zeev, Ebert Benjamin L., Chiosis Gabriela, Nimer Stephen D., Bernstein Bradley E., Verstovsek Srdan, Levine Ross L., Heterodimeric JAK–STAT activation as a mechanism of persistence to JAK2 inhibitor therapy, 10.1038/nature11303
- Fleischmann Roy, Tofacitinib in the treatment of active rheumatoid arthritis in adults, 10.2217/imt-2017-0118
- Zhang Q., Zhang Y., Diamond S., Boer J., Harris J. J., Li Y., Rupar M., Behshad E., Gardiner C., Collier P., Liu P., Burn T., Wynn R., Hollis G., Yeleswaram S., The Janus Kinase 2 Inhibitor Fedratinib Inhibits Thiamine Uptake: A Putative Mechanism for the Onset of Wernicke's Encephalopathy, 10.1124/dmd.114.058883
- Tefferi A., JAK inhibitors for myeloproliferative neoplasms: clarifying facts from myths, 10.1182/blood-2011-11-395228
- Kleppe Maria, Spitzer Matthew H., Li Sheng, Hill Corinne E., Dong Lauren, Papalexi Efthymia, De Groote Sofie, Bowman Robert L., Keller Matthew, Koppikar Priya, Rapaport Franck T., Teruya-Feldstein Julie, Gandara Jorge, Mason Christopher E., Nolan Garry P., Levine Ross L., Jak1 Integrates Cytokine Sensing to Regulate Hematopoietic Stem Cell Function and Stress Hematopoiesis, 10.1016/j.stem.2017.08.011
- Schwartz Daniella M., Bonelli Michael, Gadina Massimo, O'Shea John J., Type I/II cytokines, JAKs and new strategies for treating autoimmune diseases, 10.1038/nrrheum.2015.167
- Tefferi Ayalew, Challenges Facing JAK Inhibitor Therapy for Myeloproliferative Neoplasms, 10.1056/nejme1115119
- Bose Prithviraj, Verstovsek Srdan, JAK2 inhibitors for myeloproliferative neoplasms: what is next?, 10.1182/blood-2017-04-742288
- Leroy E, Constantinescu S N, Rethinking JAK2 inhibition: towards novel strategies of more specific and versatile janus kinase inhibition, 10.1038/leu.2017.43
- Choi J., Ziga E. D., Ritchey J., Collins L., Prior J. L., Cooper M. L., Piwnica-Worms D., DiPersio J. F., IFN R signaling mediates alloreactive T-cell trafficking and GVHD, 10.1182/blood-2012-01-403196
- Clark James D., Flanagan Mark E., Telliez Jean-Baptiste, Discovery and Development of Janus Kinase (JAK) Inhibitors for Inflammatory Diseases : Miniperspective, 10.1021/jm401490p
- Plo Isabelle, Zhang Yanyan, Le Couédic Jean-Pierre, Nakatake Mayuka, Boulet Jean-Michel, Itaya Miki, Smith Steven O., Debili Najet, Constantinescu Stefan N., Vainchenker William, Louache Fawzia, de Botton Stéphane, An activating mutation in theCSF3Rgene induces a hereditary chronic neutrophilia, 10.1084/jem.20090693
- Hong Wan-Jen, Gotlib Jason, Hereditary erythrocytosis, thrombocytosis and neutrophilia, 10.1016/j.beha.2014.07.002
- Plo Isabelle, Bellanné-Chantelot Christine, Mosca Matthieu, Mazzi Stefania, Marty Caroline, Vainchenker William, Genetic Alterations of the Thrombopoietin/MPL/JAK2 Axis Impacting Megakaryopoiesis, 10.3389/fendo.2017.00234
- Ngo Vu N., Young Ryan M., Schmitz Roland, Jhavar Sameer, Xiao Wenming, Lim Kian-Huat, Kohlhammer Holger, Xu Weihong, Yang Yandan, Zhao Hong, Shaffer Arthur L., Romesser Paul, Wright George, Powell John, Rosenwald Andreas, Muller-Hermelink Hans Konrad, Ott German, Gascoyne Randy D., Connors Joseph M., Rimsza Lisa M., Campo Elias, Jaffe Elaine S., Delabie Jan, Smeland Erlend B., Fisher Richard I., Braziel Rita M., Tubbs Raymond R., Cook J. R., Weisenburger Denny D., Chan Wing C., Staudt Louis M., Oncogenically active MYD88 mutations in human lymphoma, 10.1038/nature09671
- Lee Seungbok, Park Ha Young, Kang So Young, Kim Seok Jin, Hwang Jinha, Lee Seungho, Kwak Soo Heon, Park Kyong Soo, Yoo Hae Yong, Kim Won Seog, Kim Jong-Il, Ko Young Hyeh, Genetic alterations of JAK/STAT cascade and histone modification in extranodal NK/T-cell lymphoma nasal type, 10.18632/oncotarget.3776
- Bellanger D, Jacquemin V, Chopin M, Pierron G, Bernard O A, Ghysdael J, Stern M-H, Recurrent JAK1 and JAK3 somatic mutations in T-cell prolymphocytic leukemia, 10.1038/leu.2013.271
- Van Roosbroeck K., Cox L., Tousseyn T., Lahortiga I., Gielen O., Cauwelier B., De Paepe P., Verhoef G., Marynen P., Vandenberghe P., De Wolf-Peeters C., Cools J., Wlodarska I., JAK2 rearrangements, including the novel SEC31A-JAK2 fusion, are recurrent in classical Hodgkin lymphoma, 10.1182/blood-2010-06-291310
- Gunawardana Jay, Chan Fong Chun, Telenius Adèle, Woolcock Bruce, Kridel Robert, Tan King L, Ben-Neriah Susana, Mottok Anja, Lim Raymond S, Boyle Merrill, Rogic Sanja, Rimsza Lisa M, Guiter Chrystelle, Leroy Karen, Gaulard Philippe, Haioun Corinne, Marra Marco A, Savage Kerry J, Connors Joseph M, Shah Sohrab P, Gascoyne Randy D, Steidl Christian, Recurrent somatic mutations of PTPN1 in primary mediastinal B cell lymphoma and Hodgkin lymphoma, 10.1038/ng.2900
- Rui Lixin, Emre N.C. Tolga, Kruhlak Michael J., Chung Hye-Jung, Steidl Christian, Slack Graham, Wright George W., Lenz Georg, Ngo Vu N., Shaffer Arthur L., Xu Weihong, Zhao Hong, Yang Yandan, Lamy Laurence, Davis R. Eric, Xiao Wenming, Powell John, Maloney David, Thomas Craig J., Möller Peter, Rosenwald Andreas, Ott German, Muller-Hermelink Hans Konrad, Savage Kerry, Connors Joseph M., Rimsza Lisa M., Campo Elias, Jaffe Elaine S., Delabie Jan, Smeland Erlend B., Weisenburger Dennis D., Chan Wing C., Gascoyne Randy D., Levens David, Staudt Louis M., Cooperative Epigenetic Modulation by Cancer Amplicon Genes, 10.1016/j.ccr.2010.11.013
- Neumann Martin, Greif Philipp A., Baldus Claudia D., Mutational landscape of adult ETP-ALL, 10.18632/oncotarget.1106
- Iacobucci Ilaria, Li Yongjin, Roberts Kathryn G., Dobson Stephanie M., Kim Jaeseung C., Payne-Turner Debbie, Harvey Richard C., Valentine Marcus, McCastlain Kelly, Easton John, Yergeau Donald, Janke Laura J., Shao Ying, Chen I-Ming L., Rusch Michael, Zandi Sasan, Kornblau Steven M., Konopleva Marina, Jabbour Elias, Paietta Elisabeth M., Rowe Jacob M., Pui Ching-Hon, Gastier-Foster Julie, Gu Zhaohui, Reshmi Shalini, Loh Mignon L., Racevskis Janis, Tallman Martin S., Wiernik Peter H., Litzow Mark R., Willman Cheryl L., McPherson John D., Downing James R., Zhang Jinghui, Dick John E., Hunger Stephen P., Mullighan Charles G., Truncating Erythropoietin Receptor Rearrangements in Acute Lymphoblastic Leukemia, 10.1016/j.ccell.2015.12.013
- Shochat Chen, Tal Noa, Bandapalli Obul R., Palmi Chiara, Ganmore Ithamar, te Kronnie Geertruy, Cario Gunnar, Cazzaniga Giovanni, Kulozik Andreas E., Stanulla Martin, Schrappe Martin, Biondi Andrea, Basso Giuseppe, Bercovich Dani, Muckenthaler Martina U., Izraeli Shai, Gain-of-function mutations ininterleukin-7 receptor-α(IL7R) in childhood acute lymphoblastic leukemias, 10.1084/jem.20110580
- Hertzberg L., Vendramini E., Ganmore I., Cazzaniga G., Schmitz M., Chalker J., Shiloh R., Iacobucci I., Shochat C., Zeligson S., Cario G., Stanulla M., Strehl S., Russell L. J., Harrison C. J., Bornhauser B., Yoda A., Rechavi G., Bercovich D., Borkhardt A., Kempski H., te Kronnie G., Bourquin J.-P., Domany E., Izraeli S., Down syndrome acute lymphoblastic leukemia, a highly heterogeneous disease in which aberrant expression of CRLF2 is associated with mutated JAK2: a report from the International BFM Study Group, 10.1182/blood-2009-08-235408
- Tasian Sarah, Loh Mignon L., Understanding the Biology of CRLF2-Overexpressing Acute Lymphoblastic Leukemia, 10.1615/critrevoncog.v16.i1-2.30
- Hornakova Tekla, Staerk Judith, Royer Yohan, Flex Elisabetta, Tartaglia Marco, Constantinescu Stefan N., Knoops Laurent, Renauld Jean-Christophe, Acute Lymphoblastic Leukemia-associated JAK1 Mutants Activate the Janus Kinase/STAT Pathway via Interleukin-9 Receptor α Homodimers, 10.1074/jbc.m807531200
- Flex Elisabetta, Petrangeli Valentina, Stella Lorenzo, Chiaretti Sabina, Hornakova Tekla, Knoops Laurent, Ariola Cristina, Fodale Valentina, Clappier Emmanuelle, Paoloni Francesca, Martinelli Simone, Fragale Alessandra, Sanchez Massimo, Tavolaro Simona, Messina Monica, Cazzaniga Giovanni, Camera Andrea, Pizzolo Giovanni, Tornesello Assunta, Vignetti Marco, Battistini Angela, Cavé Hélène, Gelb Bruce D., Renauld Jean-Christophe, Biondi Andrea, Constantinescu Stefan N., Foà Robin, Tartaglia Marco, Somatically acquiredJAK1mutations in adult acute lymphoblastic leukemia, 10.1084/jem.20072182
- Malinge S., Ben-Abdelali R., Settegrana C., Radford-Weiss I., Debre M., Beldjord K., Macintyre E. A., Villeval J.-L., Vainchenker W., Berger R., Bernard O. A., Delabesse E., Penard-Lacronique V., Novel activating JAK2 mutation in a patient with Down syndrome and B-cell precursor acute lymphoblastic leukemia, 10.1182/blood-2006-09-045963
- Mullighan C. G., Zhang J., Harvey R. C., Collins-Underwood J. R., Schulman B. A., Phillips L. A., Tasian S. K., Loh M. L., Su X., Liu W., Devidas M., Atlas S. R., Chen I-M., Clifford R. J., Gerhard D. S., Carroll W. L., Reaman G. H., Smith M., Downing J. R., Hunger S. P., Willman C. L., JAK mutations in high-risk childhood acute lymphoblastic leukemia, 10.1073/pnas.0811761106
- Mullighan Charles G, Collins-Underwood J Racquel, Phillips Letha A A, Loudin Michael G, Liu Wei, Zhang Jinghui, Ma Jing, Coustan-Smith Elaine, Harvey Richard C, Willman Cheryl L, Mikhail Fady M, Meyer Julia, Carroll Andrew J, Williams Richard T, Cheng Jinjun, Heerema Nyla A, Basso Giuseppe, Pession Andrea, Pui Ching-Hon, Raimondi Susana C, Hunger Stephen P, Downing James R, Carroll William L, Rabin Karen R, Rearrangement of CRLF2 in B-progenitor– and Down syndrome–associated acute lymphoblastic leukemia, 10.1038/ng.469
- C Mullighan, Hematology Am Soc Hematol Educ Program., 2012, 389-96 (2012)
- Malinge S., Ragu C., Della-Valle V., Pisani D., Constantinescu S. N., Perez C., Villeval J.-L., Reinhardt D., Landman-Parker J., Michaux L., Dastugue N., Baruchel A., Vainchenker W., Bourquin J.-P., Penard-Lacronique V., Bernard O. A., Activating mutations in human acute megakaryoblastic leukemia, 10.1182/blood-2008-01-136366
- Yoshida Kenichi, Toki Tsutomu, Okuno Yusuke, Kanezaki Rika, Shiraishi Yuichi, Sato-Otsubo Aiko, Sanada Masashi, Park Myoung-ja, Terui Kiminori, Suzuki Hiromichi, Kon Ayana, Nagata Yasunobu, Sato Yusuke, Wang RuNan, Shiba Norio, Chiba Kenichi, Tanaka Hiroko, Hama Asahito, Muramatsu Hideki, Hasegawa Daisuke, Nakamura Kazuhiro, Kanegane Hirokazu, Tsukamoto Keiko, Adachi Souichi, Kawakami Kiyoshi, Kato Koji, Nishimura Ryosei, Izraeli Shai, Hayashi Yasuhide, Miyano Satoru, Kojima Seiji, Ito Etsuro, Ogawa Seishi, The landscape of somatic mutations in Down syndrome–related myeloid disorders, 10.1038/ng.2759
- Patnaik Mrinal M., Tefferi Ayalew, Refractory anemia with ring sideroblasts (RARS) and RARS with thrombocytosis (RARS-T): 2017 update on diagnosis, risk-stratification, and management : PATNAIK and TEFFERI, 10.1002/ajh.24637
- Maxson Julia E., Gotlib Jason, Pollyea Daniel A., Fleischman Angela G., Agarwal Anupriya, Eide Christopher A., Bottomly Daniel, Wilmot Beth, McWeeney Shannon K., Tognon Cristina E., Pond J. Blake, Collins Robert H., Goueli Basem, Oh Stephen T., Deininger Michael W., Chang Bill H., Loriaux Marc M., Druker Brian J., Tyner Jeffrey W., Oncogenic CSF3R Mutations in Chronic Neutrophilic Leukemia and Atypical CML, 10.1056/nejmoa1214514
- Reiter Andreas, Walz Christoph, Watmore Ann, Schoch Claudia, Blau Ilona, Schlegelberger Brigitte, Berger Ute, Telford Nick, Aruliah Shilani, Yin John A., Vanstraelen Danny, Barker Helen F., Taylor Peter C., O'Driscoll Aisling, Benedetti Fabio, Rudolph Cornelia, Kolb Hans-Jochem, Hochhaus Andreas, Hehlmann Rüdiger, Chase Andrew, Cross Nicholas C.P., The t(8;9)(p22;p24) Is a Recurrent Abnormality in Chronic and Acute Leukemia that FusesPCM1toJAK2, 10.1158/0008-5472.can-04-4263
- P Peeters, Blood., 90, 2535-40 (1997)
- Mark Hon Fong L., Sotomayor Edgar A., Nelson Marilu, Chaves Fernando, Sanger Warren G., Kaleem Zahid, Caughron Samuel K., Chronic idiopathic myelofibrosis (CIMF) resulting from a unique 3;9 translocation disrupting the janus kinase 2 (JAK2) gene, 10.1016/j.yexmp.2006.07.004
- Griesinger Frank, Hennig Heike, Hillmer Frauke, Podleschny Martina, Steffens Rainer, Pies Andreas, Wörmann Bernhard, Haase Detlef, Bohlander Stefan K., ABCR-JAK2 fusion gene as the result of a t(9;22)(p24;q11.2) translocation in a patient with a clinically typical chronic myeloid leukemia, 10.1002/gcc.20235
- Tefferi Ayalew, Vannucchi Alessandro Maria, Genetic Risk Assessment in Myeloproliferative Neoplasms, 10.1016/j.mayocp.2017.06.002
- Guglielmelli P, Lasho T L, Rotunno G, Score J, Mannarelli C, Pancrazzi A, Biamonte F, Pardanani A, Zoi K, Reiter A, Duncombe A, Fanelli T, Pietra D, Rumi E, Finke C, Gangat N, Ketterling R P, Knudson R A, Hanson C A, Bosi A, Pereira A, Manfredini R, Cervantes F, Barosi G, Cazzola M, Cross N C P, Vannucchi A M, Tefferi A, The number of prognostically detrimental mutations and prognosis in primary myelofibrosis: an international study of 797 patients, 10.1038/leu.2014.76
- Shimizu Takafumi, Kubovcakova Lucia, Nienhold Ronny, Zmajkovic Jakub, Meyer Sara C., Hao-Shen Hui, Geier Florian, Dirnhofer Stephan, Guglielmelli Paola, Vannucchi Alessandro M., Feenstra Jelena D. Milosevic, Kralovics Robert, Orkin Stuart H., Skoda Radek C., Loss ofEzh2synergizes withJAK2-V617F in initiating myeloproliferative neoplasms and promoting myelofibrosis, 10.1084/jem.20151136
- Ortmann Christina A., Kent David G., Nangalia Jyoti, Silber Yvonne, Wedge David C., Grinfeld Jacob, Baxter E. Joanna, Massie Charles E., Papaemmanuil Elli, Menon Suraj, Godfrey Anna L., Dimitropoulou Danai, Guglielmelli Paola, Bellosillo Beatriz, Besses Carles, Döhner Konstanze, Harrison Claire N., Vassiliou George S., Vannucchi Alessandro, Campbell Peter J., Green Anthony R., Effect of Mutation Order on Myeloproliferative Neoplasms, 10.1056/nejmoa1412098
- Saint-Martin C., Leroy G., Delhommeau F., Panelatti G., Dupont S., James C., Plo I., Bordessoule D., Chomienne C., Delannoy A., Devidas A., Gardembas-Pain M., Isnard F., Plumelle Y., Bernard O., Vainchenker W., Najman A., Bellanne-Chantelot C., , Analysis of the Ten-Eleven Translocation 2 (TET2) gene in familial myeloproliferative neoplasms, 10.1182/blood-2009-01-197525
- Delhommeau François, Dupont Sabrina, Valle Véronique Della, James Chloé, Trannoy Severine, Massé Aline, Kosmider Olivier, Le Couedic Jean-Pierre, Robert Fabienne, Alberdi Antonio, Lécluse Yann, Plo Isabelle, Dreyfus François J., Marzac Christophe, Casadevall Nicole, Lacombe Catherine, Romana Serge P., Dessen Philippe, Soulier Jean, Viguié Franck, Fontenay Michaela, Vainchenker William, Bernard Olivier A., Mutation inTET2in Myeloid Cancers, 10.1056/nejmoa0810069
- Viny Aaron D., Levine Ross L., Genetics of Myeloproliferative Neoplasms : , 10.1097/ppo.0000000000000013
- Nangalia Jyoti, Grinfeld Jacob, Green Anthony R., Pathogenesis of Myeloproliferative Disorders, 10.1146/annurev-pathol-012615-044454
- Rumi Elisa, Harutyunyan Ashot S., Pietra Daniela, Feenstra Jelena D. Milosevic, Cavalloni Chiara, Roncoroni Elisa, Casetti Ilaria, Bellini Marta, Milanesi Chiara, Renna Maria C., Gotti Manuel, Astori Cesare, Kralovics Robert, Cazzola Mario,
LNK
mutations in familial myeloproliferative neoplasms
, 10.1182/blood-2016-04-711150
- Lasho Terra L., Pardanani Animesh, Tefferi Ayalew, LNKMutations inJAK2Mutation–Negative Erythrocytosis, 10.1056/nejmc1006966
- Oh S. T., Simonds E. F., Jones C., Hale M. B., Goltsev Y., Gibbs K. D., Merker J. D., Zehnder J. L., Nolan G. P., Gotlib J., Novel mutations in the inhibitory adaptor protein LNK drive JAK-STAT signaling in patients with myeloproliferative neoplasms, 10.1182/blood-2010-02-270108
- Lu Xiaohui, Huang Lily Jun-Shen, Lodish Harvey F., Dimerization by a Cytokine Receptor Is Necessary for Constitutive Activation of JAK2V617F, 10.1074/jbc.m707125200
- Araki M., Yang Y., Masubuchi N., Hironaka Y., Takei H., Morishita S., Mizukami Y., Kan S., Shirane S., Edahiro Y., Sunami Y., Ohsaka A., Komatsu N., Activation of the thrombopoietin receptor by mutant calreticulin in CALR-mutant myeloproliferative neoplasms, 10.1182/blood-2015-09-671172
- Elf S., Abdelfattah N. S., Chen E., Perales-Paton J., Rosen E. A., Ko A., Peisker F., Florescu N., Giannini S., Wolach O., Morgan E. A., Tothova Z., Losman J.-A., Schneider R. K., Al-Shahrour F., Mullally A., Mutant Calreticulin Requires Both Its Mutant C-terminus and the Thrombopoietin Receptor for Oncogenic Transformation, 10.1158/2159-8290.cd-15-1434
- Marty C., Pecquet C., Nivarthi H., El-Khoury M., Chachoua I., Tulliez M., Villeval J.-L., Raslova H., Kralovics R., Constantinescu S. N., Plo I., Vainchenker W., Calreticulin mutants in mice induce an MPL-dependent thrombocytosis with frequent progression to myelofibrosis, 10.1182/blood-2015-11-679571
- Chachoua I., Pecquet C., El-Khoury M., Nivarthi H., Albu R.-I., Marty C., Gryshkova V., Defour J.-P., Vertenoeil G., Ngo A., Koay A., Raslova H., Courtoy P. J., Choong M. L., Plo I., Vainchenker W., Kralovics R., Constantinescu S. N., Thrombopoietin receptor activation by myeloproliferative neoplasm associated calreticulin mutants, 10.1182/blood-2015-11-681932
- Nangalia J., Massie C.E., Baxter E.J., Nice F.L., Gundem G., Wedge D.C., Avezov E., Li J., Kollmann K., Kent D.G., Aziz A., Godfrey A.L., Hinton J., Martincorena I., Van Loo P., Jones A.V., Guglielmelli P., Tarpey P., Harding H.P., Fitzpatrick J.D., Goudie C.T., Ortmann C.A., Loughran S.J., Raine K., Jones D.R., Butler A.P., Teague J.W., O'Meara S., McLaren S., Bianchi M., Silber Y., Dimitropoulou D., Bloxham D., Mudie L., Maddison M., Robinson B., Keohane C., Maclean C., Hill K., Orchard K., Tauro S., Du M.-Q., Greaves M., Bowen D., Huntly B.J.P., Harrison C.N., Cross N.C.P., Ron D., Vannucchi A.M., Papaemmanuil E., Campbell P.J., Green A.R., Somatic CALR Mutations in Myeloproliferative Neoplasms with Nonmutated JAK2, 10.1056/nejmoa1312542
- Klampfl Thorsten, Gisslinger Heinz, Harutyunyan Ashot S., Nivarthi Harini, Rumi Elisa, Milosevic Jelena D., Them Nicole C.C., Berg Tiina, Gisslinger Bettina, Pietra Daniela, Chen Doris, Vladimer Gregory I., Bagienski Klaudia, Milanesi Chiara, Casetti Ilaria Carola, Sant'Antonio Emanuela, Ferretti Virginia, Elena Chiara, Schischlik Fiorella, Cleary Ciara, Six Melanie, Schalling Martin, Schönegger Andreas, Bock Christoph, Malcovati Luca, Pascutto Cristiana, Superti-Furga Giulio, Cazzola Mario, Kralovics Robert, Somatic Mutations of Calreticulin in Myeloproliferative Neoplasms, 10.1056/nejmoa1311347
- Vainchenker William, Kralovics Robert, Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms, 10.1182/blood-2016-10-695940
- Andraos Rita, Qian Zhiyan, Bonenfant Débora, Rubert Joëlle, Vangrevelinghe Eric, Scheufler Clemens, Marque Fanny, Régnier Catherine H., De Pover Alain, Ryckelynck Hugues, Bhagwat Neha, Koppikar Priya, Goel Aviva, Wyder Lorenza, Tavares Gisele, Baffert Fabienne, Pissot-Soldermann Carole, Manley Paul W., Gaul Christoph, Voshol Hans, Levine Ross L., Sellers William R., Hofmann Francesco, Radimerski Thomas, Modulation of Activation-Loop Phosphorylation by JAK Inhibitors Is Binding Mode Dependent, 10.1158/2159-8290.cd-11-0324
- Duan Yankun, Chen Lin, Chen Yongheng, Fan Xue-gong, c-Src Binds to the Cancer Drug Ruxolitinib with an Active Conformation, 10.1371/journal.pone.0106225
- S Verstovsek, 128, 1936 (2016)
- Royer Yohan, Staerk Judith, Costuleanu Marcel, Courtoy Pierre J., Constantinescu Stefan N., Janus Kinases Affect Thrombopoietin Receptor Cell Surface Localization and Stability, 10.1074/jbc.m501376200
- Silvennoinen Olli, Ungureanu Daniela, Niranjan Yashavanthi, Hammaren Henrik, Bandaranayake Rajintha, Hubbard Stevan R., New insights into the structure and function of the pseudokinase domain in JAK2, 10.1042/bst20130005
- Toms Angela V, Deshpande Anagha, McNally Randall, Jeong Youngjee, Rogers Julia M, Kim Chae Un, Gruner Sol M, Ficarro Scott B, Marto Jarrod A, Sattler Martin, Griffin James D, Eck Michael J, Structure of a pseudokinase-domain switch that controls oncogenic activation of Jak kinases, 10.1038/nsmb.2673
- Leroy E., Dusa A., Colau D., Motamedi A., Cahu X., Mouton C., Huang L. J., Shiau A. K., Constantinescu S. N., Uncoupling JAK2 V617F activation from cytokine-induced signalling by modulation of JH2 C helix, 10.1042/bcj20160085
- Ferrao Ryan, Lupardus Patrick J., The Janus Kinase (JAK) FERM and SH2 Domains: Bringing Specificity to JAK–Receptor Interactions, 10.3389/fendo.2017.00071
- Huang Lily Jun-shen, Constantinescu Stefan N., Lodish Harvey F., The N-Terminal Domain of Janus Kinase 2 Is Required for Golgi Processing and Cell Surface Expression of Erythropoietin Receptor, 10.1016/s1097-2765(01)00401-4
- Vainchenker William, Constantinescu Stefan N., Plo Isabelle, Recent advances in understanding myelofibrosis and essential thrombocythemia, 10.12688/f1000research.8081.1
- Rampal R., Al-Shahrour F., Abdel-Wahab O., Patel J. P., Brunel J.-P., Mermel C. H., Bass A. J., Pretz J., Ahn J., Hricik T., Kilpivaara O., Wadleigh M., Busque L., Gilliland D. G., Golub T. R., Ebert B. L., Levine R. L., Integrated genomic analysis illustrates the central role of JAK-STAT pathway activation in myeloproliferative neoplasm pathogenesis, 10.1182/blood-2014-02-554634
- Vainchenker W, Constantinescu S N, JAK/STAT signaling in hematological malignancies, 10.1038/onc.2012.347
- Constantinescu Stefan N., Ghaffari Saghi, Lodish Harvey F., The Erythropoietin Receptor: Structure, Activation and Intracellular Signal Transduction, 10.1016/s1043-2760(98)00101-5
- Ihle J, TheJanusprotein tyrosine kinases in hematopoietic cytokine signaling, 10.1006/smim.1995.0029
Bibliographic reference |
William Vainchenker ; Leroy, Emilie ; Laure Gilles ; Caroline Marty ; Isabelle Plo ; et. al. JAK inhibitors for the treatment of myeloproliferative neoplasms and other disorders. In: F1000Research, Vol. 7, p. - (2018) |
Permanent URL |
http://hdl.handle.net/2078.1/193964 |