Variable IPSS DIPSS DIPSS-Plus

Total Page:16

File Type:pdf, Size:1020Kb

Variable IPSS DIPSS DIPSS-Plus Supplemental file 1: Prognostic models for myelofibrosis Prognostic models for primary myelofibrosis [1-5] Variable IPSS DIPSS DIPSS-plus Age > 65 1 1 1 Constitutional 1 1 1 Symptoms Hb < 10g/dL 1 2 1 WBC > 25 x 109/L 1 1 1 PB blasts 1% or more 1 1 1 Platelet < 100 x 109/L - - 1 RBC transfusion need - - 1 Unfavourable - - 1 karyotypeA IPSS: international prognostic scoring system; DIPSS: dynamic international prognostic scoring system; Hb: Haemoglobin; WBC: white blood cell; PB: peripheral; RBC: red blood cell. A: Unfavourable karyotype comprised +8, -7/7q-, i(17q), inv(3), -5/5q-, 12p- and 11q23 rearrangements Overall survivals predicted by prognostic models in primary myelofibrosis [1-5] Prognostic System Risk Score Median OS (months) Lille scoreA Low 0 93 Intermediate 1 26 High 2 13 IPSS Low 0 135 Int-1 1 95 Int-2 2 48 High >3 27 DIPSS Low 0 NR Int-1 1-2 170 Int-2 3-4 48 High 5-6 18 DIPSS-plus Low 0 185 Int-1 1 78 Int-2 2-3 35 High 4-6 16 IPSS: international prognostic scoring system; DIPSS: dynamic international prognostic scoring system; OS: overall survival A: Haemoglobin <10 g/dL and leucocyte count < 4 or > 30 x 109 are the two adverse prognostic indicators with a score of 1 for each. Prognostic models integrating clinical risk variable, cytogenetics and gene mutations in PMF [6-14] Variable MIPSS70 MIPSS70+ MIPSS70+ GIPSS v2.0 Anaemia 1 (Hb 1 (Hb < 2 (Hb < <10g/dL) 10g/dL) 9g/dL in men, < 8g/dL in women) 1 (Hb 9- 10.9g/dL in men, 8- 9.9g/dL in women) WBC > 25 x 109/L 2 - - - Platelet < 100 x 109/L 2 - - - Circulating blasts ≥ 2% 1 1 1 - BM fibrosis grade ≥ 2 1 - - - Constitutional symptoms 1 1 2 - Absence of type 1/type 1-like 1 2 2 1 CALR mutations Presence of an HMR mutation 1 1 2 1 (ASXL1), 1(SRSF2), 1 (U2AF1Q157) Presence of ≥2 or more HMR 2 2 3 - mutationsA Unfavourable karyotypeB - 3 3 1 Very high risk karyotypeB - - 4 2 MIPSS: mutation-enhanced international prognostic scoring system for transplant-age patients; v2.0: Version 2.0; GIPSS: genetically inspired prognosic scoring system; Hb: haemoglobin; WBC: white blood cell; BM: bone marrow; HMR: high-molecular risk. A: HMR mutations comprise ASXL1, EZH2, IDH1/2, SRFS2 and, in addition, U2AF1Q157 for MIPSS70+ version 2.0. B: Unfavourable karyotype and very high risk karyotype was defined using the revised cytogenetic risk stratification for primary myelofibrosis [13] Overall survival predicted by prognostic models integrating gene mutations in PMF [6-14] Prognostic System Risk Score Median OS (years) MIPSS70 (3-tiered) Low 0-1 Not reached Intermediate 2-4 6.3 High ≥5 3.1 MIPSS70-plus version 2.0 Very low 0 Not reached (5-tiered) Low 1-2 16.4 Intermediate 3-4 7.7 High 5-8 4.8 Very high ≥9 1.8 GIPSS (4-tiered) Low 0 26.4 Int-1 1 8 Int-2 2 4.2 High ≥3 2 MIPSS: mutation-enhanced international prognostic scoring system for transplant-age patients; v2.0: Version 2.0; GIPSS: genetically inspired prognostic scoring system; OS: overall survival References: 1. Cervantes, F.; Dupriez, B.; Pereira, A.; Passamonti, F.; Reilly, J. T.; Morra, E.; Vannucchi, A. M.; Mesa, R. A.; Demory, J. L.; Barosi, G.; Rumi, E.; Tefferi, A., New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. Blood 2009, 113, (13), 2895- 901. 2. Passamonti, F.; Cervantes, F.; Vannucchi, A. M.; Morra, E.; Rumi, E.; Pereira, A.; Guglielmelli, P.; Pungolino, E.; Caramella, M.; Maffioli, M.; Pascutto, C.; Lazzarino, M.; Cazzola, M.; Tefferi, A., A dynamic prognostic model to predict survival in primary myelofibrosis: a study by the IWG-MRT (International Working Group for Myeloproliferative Neoplasms Research and Treatment). Blood 2010, 115, (9), 1703-8. 3. Gangat, N.; Caramazza, D.; Vaidya, R.; George, G.; Begna, K.; Schwager, S.; Van Dyke, D.; Hanson, C.; Wu, W.; Pardanani, A.; Cervantes, F.; Passamonti, F.; Tefferi, A., DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. J Clin Oncol 2011, 29, (4), 392-7. 4. Morel, P.; Duhamel, A.; Hivert, B.; Stalniekiewicz, L.; Demory, J. L.; Dupriez, B., Identification during the follow-up of time-dependent prognostic factors for the competing risks of death and blast phase in primary myelofibrosis: a study of 172 patients. Blood 2010, 115, (22), 4350-5. 5. Rumi, E.; Cazzola, M., Diagnosis, risk stratification, and response evaluation in classical myeloproliferative neoplasms. Blood 2017, 129, (6), 680-692. 6. Guglielmelli, P.; Lasho, T. L.; Rotunno, G.; Mudireddy, M.; Mannarelli, C.; Nicolosi, M.; Pacilli, A.; Pardanani, A.; Rumi, E.; Rosti, V.; Hanson, C. A.; Mannelli, F.; Ketterling, R. P.; Gangat, N.; Rambaldi, A.; Passamonti, F.; Barosi, G.; Barbui, T.; Cazzola, M.; Vannucchi, A. M.; Tefferi, A., MIPSS70: Mutation-Enhanced International Prognostic Score System for Transplantation-Age Patients With Primary Myelofibrosis. J Clin Oncol 2018, 36, (4), 310-318. 7. 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.; Vannucchi, A. M.; Tefferi, A., The number of prognostically detrimental mutations and prognosis in primary myelofibrosis: an international study of 797 patients. Leukemia 2014, 28, (9), 1804-10. 8. Tefferi, A., Primary myelofibrosis: 2019 update on diagnosis, risk-stratification and management. American journal of hematology 2018, 93, (12), 1551-1560. 9. Rozovski, U.; Verstovsek, S.; Manshouri, T.; Dembitz, V.; Bozinovic, K.; Newberry, K.; Zhang, Y.; Bove, J. E. t.; Pierce, S.; Kantarjian, H.; Estrov, Z., An accurate, simple prognostic model consisting of age, JAK2, CALR, and MPL mutation status for patients with primary myelofibrosis. Haematologica 2017, 102, (1), 79-84. 10. Rumi, E.; Pietra, D.; Pascutto, C.; Guglielmelli, P.; Martinez-Trillos, A.; Casetti, I.; Colomer, D.; Pieri, L.; Pratcorona, M.; Rotunno, G.; Sant'Antonio, E.; Bellini, M.; Cavalloni, C.; Mannarelli, C.; Milanesi, C.; Boveri, E.; Ferretti, V.; Astori, C.; Rosti, V.; Cervantes, F.; Barosi, G.; Vannucchi, A. M.; Cazzola, M.; Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative, I., Clinical effect of driver mutations of JAK2, CALR, or MPL in primary myelofibrosis. Blood 2014, 124, (7), 1062- 9. 11. Tefferi, A.; Guglielmelli, P.; Nicolosi, M.; Mannelli, F.; Mudireddy, M.; Bartalucci, N.; Finke, C. M.; Lasho, T. L.; Hanson, C. A.; Ketterling, R. P.; Begna, K. H.; Naseema, G.; Pardanani, A.; Vannucchi, A. M., GIPSS: genetically inspired prognostic scoring system for primary myelofibrosis. Leukemia 2018, 32, (7), 1631-1642. 12. Tefferi, A.; Guglielmelli, P.; Pardanani, A.; Vannucchi, A. M., Myelofibrosis Treatment Algorithm 2018. Blood cancer journal 2018, 8, (8), 72. 13. Tefferi, A.; Nicolosi, M.; Mudireddy, M.; Lasho, T. L.; Gangat, N.; Begna, K. H.; Hanson, C. A.; Ketterling, R. P.; Pardanani, A., Revised cytogenetic risk stratification in primary myelofibrosis: analysis based on 1002 informative patients. Leukemia 2018, 32, (5), 1189-1199. 14. Tefferi, A.; Guglielmelli, P.; Lasho, T. L.; Gangat, N.; Ketterling, R. P.; Pardanani, A.; Vannucchi, A. M., MIPSS70+ Version 2.0: Mutation and Karyotype-Enhanced International Prognostic Scoring System for Primary Myelofibrosis. J Clin Oncol 2018, 36, (17), 1769-1770. Novel Therapies in CML NOTE: Hyperactivation of WNT974 MDM2i/HDM2i:MDM2i/HDM2i: JQ-1 Smo i: LSC survival Nutlin-3a, Plasma membrane Mechanism of TKI resistance Alteration in CML LSC Hh pathway ↑ TPO receptor JAK/STAT Nutlin-3a, Increased Sonidegib/ LDE225 MI-219, DS-5272 (MPL) hyperactivity MI-219, DS-5272 expression BMS-833923 + Dasatinib PPARγ agonist: PPARα agonist: ↑BCR-ABL1 Misoprostol +/- (TKI) PORCN Rosiglitazone Clofibrate , WY-14643 ATO + Peg-IFNα-2a Upregulation IL- Hh Wnt Wnt TNF 12β NK-cell mediated Modification Release from ER IL-1 AHI-1 JAKi: IFNα Upregulation Angiogenesis cytotoxicity JAK2 BCR-ABL1 Ruxolitinib +/- JAK2 JAK2 TKR (growth factor)-TK domain IL-12β PGE1 (Nilotinib/ -12β Wnt ↑PRMT5 IFNAR2 IFNAR1 IL PGE2 PPARγ agonist: Dasatinib/Bosutinib) Pioglitazone +/- TKI: Ruxolitinib JAK1 TYK2 (Ceased STAT3/5 STAT3/5 VEGFR NKG2D-L EP4 degradation (Imatinib) Imatinib, Dasatinib compleX) Transcription factor acts on DVL3 (Inhibition of Inhibition of Grb2 Smo PTCH FZD4 (in both nucleus hOCT1 (Nuclear SHP-2 SHC transcription Cytoplasm a.a. DVL PRMT5 phosphorylation) Amino acid APC Axin and cytoplasm) translocation) Down- (Mitochondria) 3’ Death 5’ BP1001 regulation receptor Clonal PPARγ STAT3i: Grb2 Charged GSK3β CK1 Activation of aminoacyl-tRNA evolution BP-5087 +/- 1 2 STAT3 STAT5 STAT3 ATO + Peg-IFNα-2a ATO + TKI Axin 1 RXR STAT5 Gab2 In K562 Increases in Imatinib (Imatinib) SOS STAT 55S uptake STAT (Accumulation) (Inhibition of RITA Tigecycline phosphorylation) Mitoribosome 39S Binding (Nuclear PJ-68 IKKγ IRF9 translocation) Cell cycling of Overcome TKI Transcription factor Growth arrest 5’ E-site P-site A-site 3’ Gli IKKα IKKβ DNA damage, additional Triggered ↑KRas Gli3R β-Catenin Degradation (Nuclear translocation) dormant LSCs resistance acquisition of mutations in production of ROS Ras Tigecycline 28S Kif7 Proliferation ATO + TKI CaSpaSe-8 CML LSC (homologous to 30S in bacteria) Sufu Binding to BCR-ABL1 Activation of Wnt/ Ca2+/ NFAT KRas Transcription factor Increases hyperactivation Myc MAX Myc MAX BCR-ABL1 Increased mRNA and Citrate Citrate p50 (Nuclear protein content (mitochondrial Block apoptosis and induce Binding Transcription factor IκBα NF-κB translocation) dysfunction, expansion of progenitor cells (Nuclear translocation) Src downregulation C82 + Nilotinib p65 RAF PKC Myc Degradation of ETC, increases Isocitrate + Apoptosis CaSpaSe-3, -6, -7 Dissociation PP2A oxidative stress) Isocitrate NADP NADP+ Pro-apoptotic signals from NF-κB (Upregulation àTKI resistant) BCR-ABL1 (e.g.
Recommended publications
  • Antineoplastic Agents
    bmchp.org | 888-566-0008 wellsense.org | 877-957-1300 Pharmacy Policy Antineoplastic Agents Policy Number: 9.700 Version Number: 2.0 Version Effective Date: 9/1/2021 Product Applicability All Plan+ Products Well Sense Health Plan Boston Medical Center HealthNet Plan New Hampshire Medicaid MassHealth- MCO MassHealth- ACO Qualified Health Plans/ConnectorCare/Employer Choice Direct Senior Care Options Note: Disclaimer and audit information is located at the end of this document. Prior Authorization Policy Products Affected: Daurismo™ (glasdegib tablet) Rubraca™ (rucaparib tablets) Erleada™ (apalutamide tablet) Sarclisa® (Isatuximab–IRFC injection) Farydak® (panobinostat capsule) Tazverik™ (tazemetostat tablet) Folotyn® (pralatrexate injection) Talzenna® (talazoparib capsule) Gleostine™ (lomustine capsule) Tibsovo® (ivosidenib tablet) Idhifa® (enasidenib mesylate tablet) Ukoniq (umbralisib tosylate tablet) Lonsurf® (tipiracil hcl and trifluridine tablet) Valchlor™ (mechlorethamine gel) Lynparza™ (olaparib capsule, and tablets) Zejula™ (niraparib capsules) Ninlaro® (ixazomib capsule) Zelboraf® (vemurafenib tablet) Odomzo® (sonidegib capsules) Zytiga® (abiraterone acetate tablet) Pomalyst® (pomalidomide capsule) + Plan refers to Boston Medical Center Health Plan, Inc. and its affiliates and subsidiaries offering health coverage plans to enrolled members. The Plan operates in Massachusetts under the trade name Boston Medical Center HealthNet Plan and in other states under the trade name Well Sense Health Plan. Antineoplastic Agents 1 of 4 The Plan may authorize coverage of the above products for members meeting the following criteria: Covered FDA approved indication Use Use supported by: o American Hospital Formulary Service Drug Information o DRUGDEX Information System o United States Pharmacopeia- Drug Information o National Comprehensive Cancer Network (categories 1,2a, and 2b) Medically accepted indications will also be considered for approval.
    [Show full text]
  • Mutation Analysis in Myeloproliferative Neoplasms AHS - M2101
    Corporate Medical Policy Mutation Analysis in Myeloproliferative Neoplasms AHS - M2101 File Name: mutation_analysis_in_myeloproliferative_neoplasms Origination: 1/1/2019 Last CAP review: 8/2021 Next CAP review: 8/2022 Last Review: 8/2021 Description of Procedure or Service Myeloproliferative neoplasms (MPN) are a heterogeneous group of clonal disorders characterized by overproduction of one or more differentiated myeloid lineages (Grinfeld, Nangalia, & Green, 2017). These include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). The majority of MPN result from somatic mutations in the 3 driver genes, JAK2, CALR, and MPL, which represent major diagnostic criteria in combination with hematologic and morphological abnormalities (Rumi & Cazzola, 2017). Related Policies: BCR-ABL 1 Testing for Chronic Myeloid Leukemia AHS-M2027 ***Note: This Medical Policy is complex and technical. For questions concerning the technical language and/or specific clinical indications for its use, please consult your physician. Policy BCBSNC will provide coverage for mutation analysis in myeloproliferative neoplasms when it is determined to be medically necessary because the medical criteria and guidelines shown below are met. Benefits Application This medical policy relates only to the services or supplies described herein. Please refer to the Member's Benefit Booklet for availability of benefits. Member's benefits may vary according to benefit design; therefore member benefit language should be reviewed before applying the terms of this medical policy. When Mutation Analysis in Myeloproliferative Neoplasms is covered 1. JAK2, CALR or MPL mutation testing is considered medically necessary for the diagnosis of patients presenting with clinical, laboratory, or pathological findings suggesting classic forms of myeloproliferative neoplasms (MPN), that is, polycythemia vera (PV), essential thrombocythemia (ET), or primary myelofibrosis (PMF) when ordered by a hematology and/or oncology specialist in the following situations: A.
    [Show full text]
  • Single Dose of the CXCR4 Antagonist BL-8040 Induces Rapid
    Published OnlineFirst August 23, 2017; DOI: 10.1158/1078-0432.CCR-16-2919 Cancer Therapy: Clinical Clinical Cancer Research Single Dose of the CXCR4 Antagonist BL-8040 Induces Rapid Mobilization for the Collection of Human CD34þ Cells in Healthy Volunteers Michal Abraham1, Yaron Pereg2, Baruch Bulvik1, Shiri Klein3, Inbal Mishalian3, Hana Wald1, Orly Eizenberg1, Katia Beider4, Arnon Nagler4, Rottem Golan2, Abi Vainstein2, Arnon Aharon2, Eithan Galun3, Yoseph Caraco5, Reuven Or6, and Amnon Peled3,4 Abstract Purpose: The potential of the high-affinity CXCR4 antagonist systemic reactions were mitigated by methylprednisolone, BL-8040 as a monotherapy-mobilizing agent and its derived paracetamol, and promethazine pretreatment. In the first part graft composition and quality were evaluated in a phase I clinical of the study, BL-8040 triggered rapid and substantial mobili- þ study in healthy volunteers (NCT02073019). zation of WBCs and CD34 cells in all tested doses. Four hours Experimental Design: The first part of the study was a ran- postdose, the count rose to a mean of 8, 37, 31, and 35 cells/mL domized, double-blind, placebo-controlled dose escalation (placebo, 0.5, 0.75, and 1 mg/kg, respectively). FACS analysis phase. The second part of the study was an open-label phase, in revealed substantial mobilization of immature dendritic, T, B, þ which 8 subjects received a single injection of BL-8040 (1 mg/kg) and NK cells. In the second part, the mean CD34 cells/kg and approximately 4 hours later underwent a standard leukapher- collected were 11.6 Â 106 cells/kg. The graft composition was esis procedure.
    [Show full text]
  • Promoter Janus Kinase 3 Proximal Characterization and Analysis Of
    The Journal of Immunology Characterization and Analysis of the Proximal Janus Kinase 3 Promoter1 Martin Aringer,2*† Sigrun R. Hofmann,2* David M. Frucht,* Min Chen,* Michael Centola,* Akio Morinobu,* Roberta Visconti,* Daniel L. Kastner,* Josef S. Smolen,† and John J. O’Shea3* Janus kinase 3 (Jak3) is a nonreceptor tyrosine kinase essential for signaling via cytokine receptors that comprise the common ␥-chain (␥c), i.e., the receptors for IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Jak3 is preferentially expressed in hemopoietic cells and is up-regulated upon cell differentiation and activation. Despite the importance of Jak3 in lymphoid development and immune function, the mechanisms that govern its expression have not been defined. To gain insight into this issue, we set out to characterize the Jak3 promoter. The 5؅-untranslated region of the Jak3 gene is interrupted by a 3515-bp intron. Upstream of this intron and the transcription initiation site, we identified an ϳ1-kb segment that exhibited lymphoid-specific promoter activity and was responsive to TCR signals. Truncation of this fragment revealed that core promoter activity resided in a 267-bp fragment that contains putative Sp-1, AP-1, Ets, Stat, and other binding sites. Mutation of the AP-1 sites significantly diminished, whereas mutation of the Ets sites abolished, the inducibility of the promoter construct. Chromatin immunoprecipitation assays showed that histone acetylation correlates with mRNA expression and that Ets-1/2 binds this region. Thus, transcription factors that bind these sites, especially Ets family members, are likely to be important regulators of Jak3 expression.
    [Show full text]
  • PI3K Inhibitors in Cancer: Clinical Implications and Adverse Effects
    International Journal of Molecular Sciences Review PI3K Inhibitors in Cancer: Clinical Implications and Adverse Effects Rosalin Mishra , Hima Patel, Samar Alanazi , Mary Kate Kilroy and Joan T. Garrett * Department of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267-0514, USA; [email protected] (R.M.); [email protected] (H.P.); [email protected] (S.A.); [email protected] (M.K.K.) * Correspondence: [email protected]; Tel.: +1-513-558-0741; Fax: +1-513-558-4372 Abstract: The phospatidylinositol-3 kinase (PI3K) pathway is a crucial intracellular signaling pathway which is mutated or amplified in a wide variety of cancers including breast, gastric, ovarian, colorectal, prostate, glioblastoma and endometrial cancers. PI3K signaling plays an important role in cancer cell survival, angiogenesis and metastasis, making it a promising therapeutic target. There are several ongoing and completed clinical trials involving PI3K inhibitors (pan, isoform-specific and dual PI3K/mTOR) with the goal to find efficient PI3K inhibitors that could overcome resistance to current therapies. This review focuses on the current landscape of various PI3K inhibitors either as monotherapy or in combination therapies and the treatment outcomes involved in various phases of clinical trials in different cancer types. There is a discussion of the drug-related toxicities, challenges associated with these PI3K inhibitors and the adverse events leading to treatment failure. In addition, novel PI3K drugs that have potential to be translated in the clinic are highlighted. Keywords: cancer; PIK3CA; resistance; PI3K inhibitors Citation: Mishra, R.; Patel, H.; Alanazi, S.; Kilroy, M.K.; Garrett, J.T.
    [Show full text]
  • Download (Pdf)
    Invivoscribe's wholly-owned Laboratories for Personalized Molecular LabPMM LLC Medicine® (LabPMM) is a network of international reference laboratories that provide the medical and pharmaceutical communities with worldwide Located in San Diego, California, USA, it holds access to harmonized and standardized clinical testing services. We view the following accreditations and certifications: internationally reproducible and concordant testing as a requirement for ISO 15189, CAP, and CLIA, and is licensed to provide diagnostic consistent stratification of patients for enrollment in clinical trials, and the laboratory services in the states of California, Florida, foundation for establishing optimized treatment schedules linked to patient’s Maryland, New York, Pennsylvania, and Rhode Island. individual profile. LabPMM provides reliable patient stratification at diagnosis LabPMM GmbH and monitoring, throughout the entire course of treatment in support of Personalized Molecular Medicine® and Personalized Based in Martinsried (Munich), Germany. It is an ISO 15189 Molecular Diagnostics®. accredited international reference laboratory. CLIA/CAP accreditation is planned. Invivoscribe currently operates four clinical laboratories to serve partners in the USA (San Diego, CA), Europe (Munich, Germany), and Asia (Tokyo, Japan and Shanghai, China). These laboratories use the same critical LabPMM 合同会社 reagents and software which are developed consistently with ISO Located in Kawasaki (Tokyo), Japan and a licensed clinical lab. 13485 design control. Our cGMP reagents, rigorous standards for assay development & validation, and testing performed consistently under ISO 15189 requirements help ensure LabPMM generates standardized and concordant test results worldwide. Invivoscribe Diagnostic Technologies (Shanghai) Co., Ltd. LabPMM is an international network of PersonalMed Laboratories® focused on molecular oncology biomarker studies. Located in Shangai, China.
    [Show full text]
  • Ruxolitinib for Symptom Control in Patients with Chronic Lymphocytic Leukaemia: a Single- Group, Phase 2 Trial
    UC Irvine UC Irvine Previously Published Works Title Ruxolitinib for symptom control in patients with chronic lymphocytic leukaemia: a single- group, phase 2 trial. Permalink https://escholarship.org/uc/item/1180x27h Journal The Lancet. Haematology, 4(2) ISSN 2352-3026 Authors Jain, Preetesh Keating, Michael Renner, Sarah et al. Publication Date 2017-02-01 DOI 10.1016/s2352-3026(16)30194-6 Peer reviewed eScholarship.org Powered by the California Digital Library University of California HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Lancet Haematol Manuscript Author . Author Manuscript Author manuscript; available in PMC 2018 February 01. Published in final edited form as: Lancet Haematol. 2017 February ; 4(2): e67–e74. doi:10.1016/S2352-3026(16)30194-6. Ruxolitinib for symptom control in patients with Chronic Lymphocytic leukemia: A Phase II Trial Preetesh Jain, M.D.,PhD1, Michael Keating, M.D.1,*, Sarah Renner, RN1, Charles Cleeland, PhD2,*, Huang Xuelin, PhD3,*, Graciela Nogueras Gonzalez, M.P.H3, David Harris, PhD1, Ping Li, PhD1, Zhiming Liu, PhD1, Ivo Veletic, PhD1, Uri Rozovski, M.D.1, Nitin Jain, M.D.1, Phillip Thompson, M.D.1, Prithviraj Bose, M.D.1, Courtney DiNardo, M.D.1, Alessandra Ferrajoli, M.D.1,*, Susan O’Brien, M.D.1,*, Jan Burger, M.D.1, William Wierda, M.D.1,*, Srdan Verstovsek, M.D.1,*, Hagop Kantarjian, M.D.1,*, and Zeev Estrov, M.D.1,* 1Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas 2Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas 3Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas Summary Background—Disease-related symptoms impair the quality of life of countless patients with chronic lymphocytic leukemia (CLL) who do not require systemic therapy.
    [Show full text]
  • IDH2 Mutations—Co-Opting Cellular Metabolism for Malignant Transformation Eytan M
    Published OnlineFirst November 9, 2015; DOI: 10.1158/1078-0432.CCR-15-0362 Molecular Pathways Clinical Cancer Research Molecular Pathways: IDH2 Mutations—Co-opting Cellular Metabolism for Malignant Transformation Eytan M. Stein Abstract Mutations in mitochondrial IDH2, one of the three isoforms function that catalyzes the conversion of alpha-ketoglutarate to of IDH, were discovered in patients with gliomas in 2009 and beta-hydroxyglutarate (2-HG). Supranormal levels of 2-HG subsequently described in acute myelogenous leukemia (AML), lead to hypermethylation of epigenetic targets and a subse- angioimmunoblastic T-cell lymphoma, chondrosarcoma, and quent block in cellular differentiation. AG-221, a small-mole- intrahepatic chloangiocarcinoma. The effects of mutations in cule inhibitor of mutant IDH2, is being explored in a phase I IDH2 on cellular metabolism, the epigenetic state of mutated clinical trial for the treatment of AML, other myeloid malig- cells, and cellular differentiation have been elucidated in vitro nancies, solid tumors, and gliomas. Clin Cancer Res; 22(1); 16–19. and in vivo.MutationsinIDH2 lead to an enzymatic gain of Ó2015 AACR. Background Interestingly, in a screen of 398 patients with AML, TET2 and IDH2 mutations were mutually exclusive, suggesting that these Isocitrate dehydrogenase (IDH) catalyzes the conversion of two mutations have a similar function. Wild-type TET2 demethy- isocitrate to alpha-ketoglutarate. IDH occurs in three isoforms, lates DNA, and mutations in TET2 lead to increases in 5-methyl- IDH1, located in the cytoplasm, IDH2 located in the mitochon- cytosine similar to those seen in patients with IDH mutations. It dria, and IDH3, which functions as part of the tricarboxylic acid has been suggested that elevated levels of 2-HG caused by mutant cycle.
    [Show full text]
  • 942.Full.Pdf
    Original Article Opposite Effect of JAK2 on Insulin-Dependent Activation of Mitogen-Activated Protein Kinases and Akt in Muscle Cells Possible Target to Ameliorate Insulin Resistance Ana C.P. Thirone, Lellean JeBailey, Philip J. Bilan, and Amira Klip Many cytokines increase their receptor affinity for Janus kinases (JAKs). Activated JAK binds to signal transducers and activators of transcription, insulin receptor substrates olypeptides such as erythropoietin, prolactin, (IRSs), and Shc. Intriguingly, insulin acting through its leptin, angiotensin, growth hormone, most inter- own receptor kinase also activates JAK2. However, the leukins, and interferon-␥ bind to receptors that impact of such activation on insulin action remains un- Plack intrinsic kinase activity, recruiting and acti- known. To determine the contribution of JAK2 to insulin vating cytoplasmic tyrosine kinases of the Janus family signaling, we transfected L6 myotubes with siRNA against (JAK) consisting of JAK1, JAK2, JAK3, and Tyk2 (1–3). JAK2 (siJAK2), reducing JAK2 protein expression by 75%. Activated JAK phosphorylates tyrosine residues within Insulin-dependent phosphorylation of IRS1/2 and Shc was not affected by siJAK2, but insulin-induced phosphoryla- itself and the associated receptor forming high-affinity tion of the mitogen-activated protein kinases (MAPKs) binding sites for a variety of signaling proteins containing Src homology 2 and other phosphotyrosine-binding do- extracellular signal–related kinase, p38, and Jun NH2- terminal kinase and their respective upstream kinases mains, including signal transducers and activators of tran- MKK1/2, MKK3/6, and MKK4/7 was significantly lowered scription, insulin receptor substrates (IRSs), and the when JAK2 was depleted, correlating with a significant adaptor protein Shc (1–4).
    [Show full text]
  • JAK Inhibitors for Treatment of Psoriasis: Focus on Selective TYK2 Inhibitors
    Drugs https://doi.org/10.1007/s40265-020-01261-8 CURRENT OPINION JAK Inhibitors for Treatment of Psoriasis: Focus on Selective TYK2 Inhibitors Miguel Nogueira1 · Luis Puig2 · Tiago Torres1,3 © Springer Nature Switzerland AG 2020 Abstract Despite advances in the treatment of psoriasis, there is an unmet need for efective and safe oral treatments. The Janus Kinase– Signal Transducer and Activator of Transcription (JAK–STAT) pathway plays a signifcant role in intracellular signalling of cytokines of numerous cellular processes, important in both normal and pathological states of immune-mediated infamma- tory diseases. Particularly in psoriasis, where the interleukin (IL)-23/IL-17 axis is currently considered the crucial pathogenic pathway, blocking the JAK–STAT pathway with small molecules would be expected to be clinically efective. However, relative non-specifcity and low therapeutic index of the available JAK inhibitors have delayed their integration into the therapeutic armamentarium of psoriasis. Current research appears to be focused on Tyrosine kinase 2 (TYK2), the frst described member of the JAK family. Data from the Phase II trial of BMS-986165—a selective TYK2 inhibitor—in psoriasis have been published and clinical results are encouraging, with a large Phase III programme ongoing. Further, the selective TYK2 inhibitor PF-06826647 is being tested in moderate-to-severe psoriasis in a Phase II clinical trial. Brepocitinib, a potent TYK2/JAK1 inhibitor, is also being evaluated, as both oral and topical treatment. Results of studies with TYK2 inhibitors will be important in assessing the clinical efcacy and safety of these drugs and their place in the therapeutic armamentarium of psoriasis.
    [Show full text]
  • Modulators of CXCR4 and CXCR7/ACKR3 Function
    Molecular Pharmacology Fast Forward. Published on September 23, 2019 as DOI: 10.1124/mol.119.117663 This article has not been copyedited and formatted. The final version may differ from this version. MOL # 117663 Modulators of CXCR4 and CXCR7/ACKR3 function Ilze Adlere*, Birgit Caspar*, Marta Arimont*, Sebastian Dekkers, Kirsten Visser, Jeffrey Stuijt, Chris de Graaf, Michael Stocks, Barrie Kellam, Stephen Briddon, Maikel Wijtmans, Iwan de Esch, Stephen Hill, Rob Leurs# * These authors contributed equally to this work. Downloaded from # Corresponding author molpharm.aspetjournals.org Griffin Discoveries BV, Amsterdam, The Netherlands (IA, IE, RL), Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK (BC, SJB, SJH), Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, U.K. (BC, BK, SD, SB, SH), at ASPET Journals on September 26, 2021 School of Pharmacy, University of Nottingham, Nottingham, U.K. (SD, BK, MS), Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (MA, KS, JS, CG, MW, IE, RL), Sosei Heptares, Cambridge, U.K. (CG) 1 Molecular Pharmacology Fast Forward. Published on September 23, 2019 as DOI: 10.1124/mol.119.117663 This article has not been copyedited and formatted. The final version may differ from this version. MOL # 117663 Running title: Modulators of CXCR4 and CXCR7/ACKR3 function Corresponding
    [Show full text]
  • Dynamic Chemotherapy-Induced Upregulation of CXCR4 Expression: a Mechanism of Therapeutic Resistance in Pediatric AML
    Published OnlineFirst June 10, 2013; DOI: 10.1158/1541-7786.MCR-13-0114 Molecular Cancer Cell Death and Survival Research Dynamic Chemotherapy-Induced Upregulation of CXCR4 Expression: A Mechanism of Therapeutic Resistance in Pediatric AML Edward Allan R. Sison, Emily McIntyre, Daniel Magoon, and Patrick Brown Abstract Cure rates in pediatric acute leukemias remain suboptimal. Overexpression of the cell-surface chemokine receptor CXCR4 is associated with poor outcome in acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). Certain nonchemotherapeutic agents have been shown to modulate CXCR4 expression and alter leukemia interactions with stromal cells in the bone marrow microenvironment. Because chemotherapy is the mainstay of AML treatment, it was hypothesized that standard cytotoxic chemotherapeutic agents induce dynamic changes in leukemia surface CXCR4 expression, and that chemotherapy-induced upregulation of CXCR4 repre- sents a mechanism of acquired therapeutic resistance. Here, it was shown that cell lines variably upregulate CXCR4 with chemotherapy treatment. Those that showed upregulation were differentially protected from chemotherapy- induced apoptosis when cocultured with stroma. The functional effects of chemotherapy-induced CXCR4 up- regulation in an AML cell line (MOLM-14, which harbors consistent upregulated CXCR4) and clinical specimens were explored. Importantly, enhanced stromal-cell derived factor-1a (SDF1A/CXCL12)-mediated chemotaxis and stromal protection from additional chemotherapy-induced apoptosis was found. Furthermore, treatment with plerixafor, a CXCR4 inhibitor, preferentially decreased stromal protection with higher chemotherapy-induced upregulation of surface CXCR4. Thus, increased chemokine receptor CXCR4 expression after treatment with conventional chemotherapy may represent a mechanism of therapeutic resistance in pediatric AML. Implications: CXCR4 may be a biomarker for the stratification and optimal treatment of patients using CXCR4 inhibitors.
    [Show full text]