1
|
Herqash RN, Alkathiri FA, Darwish IA. Assessing pharmacokinetics and drug-drug interactions of the combination therapy of myelofibrosis with ruxolitinib and lenalidomide by a new eco-friendly HPLC method for their simultaneous determination in plasma. Cancer Chemother Pharmacol 2024; 94:747-761. [PMID: 39259291 DOI: 10.1007/s00280-024-04715-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
Ruxolitinib (RUX), a Janus kinase 2 (JAK2) inhibitor, and lenalidomide (LEN), an immunomodulatory agent, have recently been proposed as a combined treatment for myelofibrosis (MF). This combination has demonstrated improved efficacy, safety, and tolerability compared to monotherapy. To further refine these findings, an efficient analytical tool is needed to simultaneously determine RUX and LEN concentrations in blood plasma. This tool would enable the study of their pharmacokinetics, drug-drug interactions, and therapeutic monitoring during MF therapy. Unfortunately, such a method has not been existed in the literature. This study presents the first HPLC method with UV detection for the simultaneous quantitation of RUX and LEN in plasma. The method was validated according to the ICH guidelines for bioanalytical method validation. It exhibited linearity in the concentration ranges of 10 to 3150 ng mL- 1 for RUX and 80 to 5200 ng mL- 1 for LEN. The limits of quantitation were determined to be 25 and 90 ng mL- 1 for RUX and LEN, respectively. All other validation parameters were satisfactory. The HPLC-UV method was successfully employed to study the pharmacokinetics and drug-drug interactions of RUX and LEN in rats following oral administration of single doses. The results demonstrated that the pharmacokinetics of both drugs were changed substantially by their coadministration. LEN exhibited synergistic effects on the maximum plasma concentration (Cmax) and total bioavailability of RUX, meanwhile it exhibited diminishing effect on the values of volume of distribution (Vd) and clearance (CL). Additionally, RUX decreased the Cmax and total bioavailability of LEN, meanwhile it increased its Vd and CL. These data suggest that the use of RUX, as a combination with LEN, is a better therapeutic approach for MF, compared with RUX as a monotherapy. The effects of LEN on the pharmacokinetics of RUX should be considered and can be useful in determining the appropriate RUX dosage and dosing regimen to achieve the desired therapeutic effect when used as a combination therapy with LEN. The method's environmental friendliness was confirmed through three comprehensive tools. This method represents a valuable tool for determining the appropriate dosage and dosing regimen of RUX in combination therapy with LEN to achieve the desired therapeutic effect. Furthermore, it can aid in predicting drug distribution in different patients and assessing the drug accumulation or insufficient drug levels in specific body compartments.
Collapse
Affiliation(s)
- Rashed N Herqash
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Fai A Alkathiri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ibrahim A Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia.
| |
Collapse
|
2
|
Pomicter AD, Clair PM, Yan D, Heaton WL, Eiring AM, Anderson MB, Richards SM, Gililland J, O'Hare T, Deininger MW. Femoral Heads from Total Hip Arthroplasty as a Source of Adult Hematopoietic Cells. Acta Haematol 2021; 144:458-464. [PMID: 33412552 DOI: 10.1159/000511953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/28/2020] [Indexed: 12/19/2022]
Abstract
Normal human bone marrow cells are critical for studies of hematopoiesis and as controls to assess toxicity. As cells from commercial vendors are expensive, many laboratories resort to cancer-free bone marrow specimens obtained during staging or to umbilical cord blood cells, which may be abnormal or reflect a much younger age group compared to the disease samples under study. We piloted the use of femoral heads as an alternative and inexpensive source of normal bone marrow. Femoral heads were obtained from 21 successive patients undergoing elective hip arthroplasty. Mononuclear cells (MNCs) were purified with Ficoll, and CD3+, CD14+, and CD34+ cells were purified with antibody-coated microbeads. The median yield of MNCs was 8.95 × 107 (range, 1.62 × 105-2.52 × 108), and the median yield of CD34+ cells was 1.40 × 106 (range, 3.60 × 105-9.90 × 106). Results of downstream applications including qRT-PCR, colony-forming assays, and ex vivo proliferation analysis were of high quality and comparable to those obtained with standard bone marrow aspirates. We conclude that femoral heads currently discarded as medical waste are a cost-efficient source of bone marrow cells for research use.
Collapse
Affiliation(s)
- Anthony D Pomicter
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Phillip M Clair
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Dongqing Yan
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - William L Heaton
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Anna M Eiring
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Michael B Anderson
- Department of Orthopaedic Surgery, University of Utah, Salt Lake City, Utah, USA
| | - Stephen M Richards
- Department of Orthopaedic Surgery, University of Utah, Salt Lake City, Utah, USA
| | - Jeremy Gililland
- Department of Orthopaedic Surgery, University of Utah, Salt Lake City, Utah, USA
| | - Thomas O'Hare
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
- Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, Utah, USA
| | - Michael W Deininger
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA,
- Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, Utah, USA,
| |
Collapse
|
3
|
Padrnos L, Mesa R. Novel agents for the treatment of polycythemia vera: an insight into preclinical research and early phase clinical trials. Expert Opin Investig Drugs 2020; 29:809-817. [DOI: 10.1080/13543784.2020.1782886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Leslie Padrnos
- Division of Hematology and Medical Oncology, Mayo Clinic , Phoenix, Arizona, USA
| | - Ruben Mesa
- Department of Hematology and Oncology, UT Health Science Center San Antonio MD Anderson Cancer Center , San Antonio, Texas, USA
| |
Collapse
|
4
|
C. Diaconu C, Gurban P, Mambet C, Chivu-Economescu M, G. Necula L, Matei L, Dragu D, Nedeianu S, I. Neagu A, Tatic A, Cristodor D, Bleotu C. Programmed Cell Death Deregulation in BCR-ABL1-Negative Myeloproliferative Neoplasms. PROGRAMMED CELL DEATH 2020. [DOI: 10.5772/intechopen.86062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
|
5
|
Zheng G, Chen P, Pallavajjalla A, Haley L, Gondek L, Dezern A, Ling H, De Marchi F, Lin MT, Gocke C. The diagnostic utility of targeted gene panel sequencing in discriminating etiologies of cytopenia. Am J Hematol 2019; 94:1141-1148. [PMID: 31350794 PMCID: PMC9162094 DOI: 10.1002/ajh.25592] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 07/15/2019] [Accepted: 07/19/2019] [Indexed: 01/03/2023]
Abstract
The diagnostic utility of somatic mutations in the context of cytopenias is unclear: clonal hematopoiesis can be found in healthy individuals, patients with aplastic anemia (AA), clonal cytopenia of undetermined significance (CCUS) and myelodysplastic syndrome (MDS). We examined a cohort of 207 well-characterized cytopenic patients with a 640-gene next generation sequencing (NGS) panel and compared its diagnostic utility with a "virtual" 41 gene panel. The TET2, SF3B1, ASXL1, and TP53 were the most commonly mutated genes (frequency > 10%). Mutations in the 640-gene panel show high sensitivity (98.3%) but low specificity (47.6%) for diagnosis of MDS. Notably, mutations of splicing factors and genes in the RAS pathway are relatively specific to MDS. Furthermore, high variant allele frequency (VAF) predicts MDS: when the VAF is set at 20%, the positive predictive value (PPV) for MDS is 95.9%, with a specificity of 95.3%. The presence of two or more somatic mutations with ≥10% VAF showed a PPV of 95.2%. While the "virtual" 41-gene panel showed a mild decrease in sensitivity (95.7% vs 98.3%), 100% specificity was observed when either VAF was set at ≥20% (100% vs 95.3%), or two or more somatic mutations had VAFs ≥ 10%. Our study shows targeted gene panel sequencing improves the diagnostic approach and accuracy for unexplained cytopenia, with its high sensitivity and high PPV for MDS when applying VAF cutoffs. Furthermore, a 41-gene panel was shown to have at least comparable performance characteristics to the large 640-gene panel.
Collapse
Affiliation(s)
- Gang Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| | - Ping Chen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Hematology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Aparna Pallavajjalla
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lisa Haley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lukasz Gondek
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Amy Dezern
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hua Ling
- Center for Inherited Disease Research, Johns Hopkins University, Baltimore, Maryland
| | - Federico De Marchi
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher Gocke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
6
|
Shimada A. Hematological malignancies and molecular targeting therapy. Eur J Pharmacol 2019; 862:172641. [PMID: 31493406 DOI: 10.1016/j.ejphar.2019.172641] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/08/2019] [Accepted: 09/02/2019] [Indexed: 01/12/2023]
Abstract
Recent genetic analysis using next-generation sequencing (NGS) vastly improved the understanding of molecular mechanism of hematological malignancies. Many molecular targeting drugs have since been used in the clinic, which is timely as clinical outcomes using conventional chemotherapy and hematopoietic stem cell transplantation (HSCT) reached a plateau. The first memorable success in this field was imatinib, a first-generation tyrosine kinase inhibitor (TKI), which has been applied in chronic myeloid leukemia (CML) since 2001. Imatinib drastically changed CML treatment and many CML patients no longer require HSCT. Recently, the second generation TKIs, dasatinib, nilotinib, and ponatinib, have also been available for CML patients. Acute lymphoblastic leukemia (ALL) is sub-categorized based on cytogenetic or molecular genetic abnormalities. Chemotherapy and HSCT combined with TKI improved the event-free survival rate from 20% to 80% in Philadelphia (Ph) chromosome-positive ALL. Reportedly, another Ph-like ALL subgroup with poor prognosis can also be treated by TKIs; additionally, cell therapies that include bispecific T-cell engagers or chimeric antigen receptor (CAR)-T therapy are emerging. Acute myeloid leukemia (AML) is a heterogenous disease and FMS-like related tyrosine kinase-3 (FLT3)-internal tandem duplication, is the most robust marker for poor prognosis. Several first-generation TKIs have been studied for clinical use. Notably, chemotherapy plus midostaurin improved survival compared with chemotherapy alone. Therefore, midostaurin was approved to treat adult AML patients with FLT3-ITD in 2017. Gemtuzumab ozogamicin, a selective anti-CD33 antibody-calicheamicin conjugate, is approved for clinical practice. Many molecular targeting agents are now being used for hematological malignancies.
Collapse
Affiliation(s)
- Akira Shimada
- Department of Pediatric Hematology and Oncology, Okayama University Hospital, Okayama, 700-8558, Japan.
| |
Collapse
|
7
|
Grunwald MR, Burke JM, Kuter DJ, Gerds AT, Stein B, Walshauser MA, Parasuraman S, Colucci P, Paranagama D, Savona MR, Mesa R. Symptom Burden and Blood Counts in Patients With Polycythemia Vera in the United States: An Analysis From the REVEAL Study. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:579-584.e1. [PMID: 31303457 PMCID: PMC8148986 DOI: 10.1016/j.clml.2019.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/14/2019] [Accepted: 06/04/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Approximately 50% of patients with polycythemia vera (PV) have PV-related symptoms at diagnosis; these symptoms might develop or worsen with time. Symptoms have been shown to negatively affect quality of life and interfere with daily activities. To our knowledge, an analysis to evaluate the relationship between blood count control and symptoms has not been published. PATIENTS AND METHODS The Prospective Observational Study of Patients with Polycythemia Vera in US Clinical Practices (REVEAL; NCT02252159) is a multicenter, noninterventional, nonrandomized prospective observational study of patients with PV in the United States. Patients included were required to have a complete blood count result within 30 days before completing the at-enrollment Myeloproliferative Neoplasm Self-Assessment Form Total Symptom Score (MPN-SAF TSS). Symptom severity was compared between those who had blood count control versus those who did not. RESULTS At the time of enrollment, 1714 patients (94.5%) were being managed with cytoreductive therapy; 468 patients (25.8%) had complete hematologic remission (CHR), 1614 patients (89.0%) had ≥1 controlled blood count, and 1122 patients (61.9%) had ≥2 controlled blood counts. Mean MPN-SAF TSSs were similar across patients in different blood count control groups. Fatigue was the most frequently reported symptom. The severity of individual symptoms, except those of pruritus and night sweats, was not affected by CHR or the number of blood counts that were controlled. CONCLUSION Symptom burden in patients with PV can persist despite control of blood counts, which suggests some discordance between laboratory values and symptom burden. Consequently, regular monitoring of symptom burden should be factored into the assessment of disease control.
Collapse
Affiliation(s)
- Michael R Grunwald
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC.
| | - John M Burke
- US Oncology Hematology Research Program, Rocky Mountain Cancer Centers, Aurora, CO
| | - David J Kuter
- Center for Hematology, Massachusetts General Hospital, Boston, MA
| | - Aaron T Gerds
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH
| | - Brady Stein
- Department of Hematology and Oncology, Northwestern University, Chicago, IL
| | - Mark A Walshauser
- Department of Medical Oncology and Hematology, Cancer Care Specialists of Illinois, Swansea, IL
| | | | | | | | - Michael R Savona
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Ruben Mesa
- Department of Hematology and Oncology, The University of Texas Health Science Center, San Antonio, TX
| |
Collapse
|
8
|
Leroy E, Balligand T, Pecquet C, Mouton C, Colau D, Shiau AK, Dusa A, Constantinescu SN. Differential effect of inhibitory strategies of the V617 mutant of JAK2 on cytokine receptor signaling. J Allergy Clin Immunol 2019; 144:224-235. [PMID: 30707971 DOI: 10.1016/j.jaci.2018.12.1023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 12/14/2018] [Accepted: 12/20/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Janus kinase (JAK) 2 plays pivotal roles in signaling by several cytokine receptors. The mutant JAK2 V617F is the most common molecular event associated with myeloproliferative neoplasms. Selective targeting of the mutant would be ideal for treating these pathologies by sparing essential JAK2 functions. OBJECTIVE We characterize inhibitory strategies for JAK2 V617F and assess their effect on physiologic signaling by distinct cytokine receptors. METHODS Through structure-guided mutagenesis, we assessed the role of key residues around F617 and used a combination of cellular and biochemical assays to measure the activity of JAKs in reconstituted cells. We also assessed the effect of several specific JAK2 V617F inhibitory mutations on receptor dimerization using the NanoBiT protein complementation approach. RESULTS We identified a novel Janus kinase homology 2 (JH2) αC mutation, A598F, which is suggested to inhibit the aromatic stacking between F617 with F594 and F595. Like other JAK2 V617F inhibitory mutations, A598F decreased oncogenic activation and spared cytokine activation while preventing JAK2 V617F-promoted erythropoietin receptor dimerization. Surprisingly, A598F and other V617F-inhibiting mutations (F595A, E596R, and F537A) significantly impaired IFN-γ signaling. This was specific for IFN-γ because the inhibitory mutations preserved responses to ligands of a series of receptor complexes. Similarly, homologous mutations in JAK1 prevented signaling by IFN-γ. CONCLUSIONS The JH2 αC region, which is required for JAK2 V617F hyperactivation, is crucial for relaying cytokine-induced signaling of the IFN-γ receptor. We discuss how strategies aiming to inhibit JAK2 V617F could be used for identifying inhibitors of IFN-γ signaling.
Collapse
Affiliation(s)
- Emilie Leroy
- Ludwig Institute for Cancer Research, Brussels, Belgium; de Duve Institute, Université Catholique de Louvain, Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium
| | - Thomas Balligand
- Ludwig Institute for Cancer Research, Brussels, Belgium; de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Christian Pecquet
- Ludwig Institute for Cancer Research, Brussels, Belgium; de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Céline Mouton
- Ludwig Institute for Cancer Research, Brussels, Belgium; de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Didier Colau
- Ludwig Institute for Cancer Research, Brussels, Belgium; de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Andrew K Shiau
- Small Discovery Program, Ludwig Institute for Cancer Research, La Jolla, Calif
| | - Alexandra Dusa
- Ludwig Institute for Cancer Research, Brussels, Belgium; de Duve Institute, Université Catholique de Louvain, Brussels, Belgium.
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research, Brussels, Belgium; de Duve Institute, Université Catholique de Louvain, Brussels, Belgium; WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium.
| |
Collapse
|
9
|
Yan D, Pomicter AD, Tantravahi S, Mason CC, Senina AV, Ahmann JM, Wang Q, Than H, Patel AB, Heaton WL, Eiring AM, Clair PM, Gantz KC, Redwine HM, Swierczek SI, Halverson BJ, Baloglu E, Shacham S, Khorashad JS, Kelley TW, Salama ME, Miles RR, Boucher KM, Prchal JT, O'Hare T, Deininger MW. Nuclear-Cytoplasmic Transport Is a Therapeutic Target in Myelofibrosis. Clin Cancer Res 2018; 25:2323-2335. [PMID: 30563936 DOI: 10.1158/1078-0432.ccr-18-0959] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/31/2018] [Accepted: 12/14/2018] [Indexed: 01/06/2023]
Abstract
PURPOSE Myelofibrosis is a hematopoietic stem cell neoplasm characterized by bone marrow reticulin fibrosis, extramedullary hematopoiesis, and frequent transformation to acute myeloid leukemia. Constitutive activation of JAK/STAT signaling through mutations in JAK2, CALR, or MPL is central to myelofibrosis pathogenesis. JAK inhibitors such as ruxolitinib reduce symptoms and improve quality of life, but are not curative and do not prevent leukemic transformation, defining a need to identify better therapeutic targets in myelofibrosis. EXPERIMENTAL DESIGN A short hairpin RNA library screening was performed on JAK2V617F-mutant HEL cells. Nuclear-cytoplasmic transport (NCT) genes including RAN and RANBP2 were among top candidates. JAK2V617F-mutant cell lines, human primary myelofibrosis CD34+ cells, and a retroviral JAK2V617F-driven myeloproliferative neoplasms mouse model were used to determine the effects of inhibiting NCT with selective inhibitors of nuclear export compounds KPT-330 (selinexor) or KPT-8602 (eltanexor). RESULTS JAK2V617F-mutant HEL, SET-2, and HEL cells resistant to JAK inhibition are exquisitely sensitive to RAN knockdown or pharmacologic inhibition by KPT-330 or KPT-8602. Inhibition of NCT selectively decreased viable cells and colony formation by myelofibrosis compared with cord blood CD34+ cells and enhanced ruxolitinib-mediated growth inhibition and apoptosis, both in newly diagnosed and ruxolitinib-exposed myelofibrosis cells. Inhibition of NCT in myelofibrosis CD34+ cells led to nuclear accumulation of p53. KPT-330 in combination with ruxolitinib-normalized white blood cells, hematocrit, spleen size, and architecture, and selectively reduced JAK2V617F-mutant cells in vivo. CONCLUSIONS Our data implicate NCT as a potential therapeutic target in myelofibrosis and provide a rationale for clinical evaluation in ruxolitinib-exposed patients with myelofibrosis.
Collapse
Affiliation(s)
- Dongqing Yan
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | | | - Srinivas Tantravahi
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah.,Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| | - Clinton C Mason
- Department of Pediatrics, The University of Utah, Salt Lake City, Utah
| | - Anna V Senina
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Jonathan M Ahmann
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Qiang Wang
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah.,Department of Hematology, Nanfang Hospital, Southern Medical University
| | - Hein Than
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah.,Department of Haematology, Singapore General Hospital, Singapore
| | - Ami B Patel
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah.,Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| | - William L Heaton
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Anna M Eiring
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Phillip M Clair
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Kevin C Gantz
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Hannah M Redwine
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Sabina I Swierczek
- Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| | | | | | | | - Jamshid S Khorashad
- Department of Cellular Pathology, Hammersmith Hospital, Imperial College Health Care NHS Trust, London, United Kingdom
| | - Todd W Kelley
- Department of Pathology, The University of Utah, Salt Lake City, Utah
| | - Mohamed E Salama
- Department of Pathology, The University of Utah, Salt Lake City, Utah
| | - Rodney R Miles
- Department of Pathology, The University of Utah, Salt Lake City, Utah
| | - Kenneth M Boucher
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah
| | - Josef T Prchal
- Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| | - Thomas O'Hare
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah.,Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| | - Michael W Deininger
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah. .,Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, Utah
| |
Collapse
|
10
|
Cardoso EM, Esgalhado AJ, Patrão L, Santos M, Neves VP, Martinez J, Patto MAV, Silva H, Arosa FA. Distinctive CD8 + T cell and MHC class I signatures in polycythemia vera patients. Ann Hematol 2018; 97:1563-1575. [PMID: 29789880 DOI: 10.1007/s00277-018-3332-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 04/05/2018] [Indexed: 12/18/2022]
Abstract
Polycythemia vera (PV) is a myeloproliferative neoplasm characterized by overproduction of red blood cells. We have performed a comprehensive characterization of blood immune cells for expression of naïve and memory receptors as well as β2m-associated and β2m-free MHC class I heavy chains, also known as closed and open conformers, respectively, in PV patients and age-matched controls (CTR). We show that the peripheral CD3+CD8+ T cell pool in PV patients is clearly divided into two discrete populations, a more granular CD3+CD8high T cell population enriched in effector-memory CD45RA+ T cells (CD8+ TEMRA) when compared to CTR (P < 0.001), and a less granular CD3+CD8int T cell population that is completely absent in the CTR group (78 vs. 0%, P < 0.001) and is a mixture of naïve (CD8+ TN) and CD8+ TEMRA cells expressing intermediate levels of CD28, i.e., CD3+CD8intCD28int. While the percentage of CD3+CD8int TN cells correlated positively with the number of erythrocytes, the percentage of CD3+CD8int TEMRA correlated negatively with the number of platelets. Finally, we report that PV patients' lymphocytes and monocytes display lower levels of closed (W6/32+) MHC-I conformers at the cell surface while exhibiting increased amounts of open (HC-10+) MHC-I conformers. The implications of this distinctive immune signature are discussed.
Collapse
Affiliation(s)
- Elsa M Cardoso
- CICS-UBI-Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal.,FCS-Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,IPG-Instituto Politécnico da Guarda, Guarda, Portugal
| | - André J Esgalhado
- CICS-UBI-Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Luís Patrão
- FCS-Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,CHTV-Centro Hospitalar Tondela-Viseu, Viseu, Portugal
| | - Mónica Santos
- CHTV-Centro Hospitalar Tondela-Viseu, Viseu, Portugal
| | | | - Jorge Martinez
- FCS-Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,CHCB-Centro Hospitalar Cova da Beira, Covilhã, Portugal
| | - Maria Assunção Vaz Patto
- CICS-UBI-Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal.,FCS-Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,ULSG-Unidade Local de Saúde, Guarda, Portugal
| | - Helena Silva
- CHTV-Centro Hospitalar Tondela-Viseu, Viseu, Portugal
| | - Fernando A Arosa
- CICS-UBI-Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal. .,FCS-Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
11
|
Lue HW, Podolak J, Kolahi K, Cheng L, Rao S, Garg D, Xue CH, Rantala JK, Tyner JW, Thornburg KL, Martinez-Acevedo A, Liu JJ, Amling CL, Truillet C, Louie SM, Anderson KE, Evans MJ, O'Donnell VB, Nomura DK, Drake JM, Ritz A, Thomas GV. Metabolic reprogramming ensures cancer cell survival despite oncogenic signaling blockade. Genes Dev 2017; 31:2067-2084. [PMID: 29138276 PMCID: PMC5733498 DOI: 10.1101/gad.305292.117] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/26/2017] [Indexed: 12/19/2022]
Abstract
Lue et al. show that although inhibition of PI3K–AKT–mTOR signaling markedly decreased glycolysis and restrained tumor growth, these signaling and metabolic restrictions triggered autophagy. Survival of cancer cells was critically dependent on phospholipase A2 (PLA2) to mobilize lysophospholipids and free fatty acids to sustain fatty acid oxidation and oxidative phosphorylation. There is limited knowledge about the metabolic reprogramming induced by cancer therapies and how this contributes to therapeutic resistance. Here we show that although inhibition of PI3K–AKT–mTOR signaling markedly decreased glycolysis and restrained tumor growth, these signaling and metabolic restrictions triggered autophagy, which supplied the metabolites required for the maintenance of mitochondrial respiration and redox homeostasis. Specifically, we found that survival of cancer cells was critically dependent on phospholipase A2 (PLA2) to mobilize lysophospholipids and free fatty acids to sustain fatty acid oxidation and oxidative phosphorylation. Consistent with this, we observed significantly increased lipid droplets, with subsequent mobilization to mitochondria. These changes were abrogated in cells deficient for the essential autophagy gene ATG5. Accordingly, inhibition of PLA2 significantly decreased lipid droplets, decreased oxidative phosphorylation, and increased apoptosis. Together, these results describe how treatment-induced autophagy provides nutrients for cancer cell survival and identifies novel cotreatment strategies to override this survival advantage.
Collapse
Affiliation(s)
- Hui-Wen Lue
- Knight Comprehensive Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Jennifer Podolak
- Knight Comprehensive Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Kevin Kolahi
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Larry Cheng
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Soumya Rao
- Knight Comprehensive Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Devin Garg
- Knight Comprehensive Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Chang-Hui Xue
- Knight Comprehensive Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Juha K Rantala
- Knight Comprehensive Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Jeffrey W Tyner
- Knight Comprehensive Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Kent L Thornburg
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Ann Martinez-Acevedo
- Department of Urology, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Jen-Jane Liu
- Department of Urology, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Christopher L Amling
- Department of Urology, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Charles Truillet
- Department of Radiology, University of California at San Francisco School of Medicine, San Francisco, California 94107, USA
| | - Sharon M Louie
- University of California at Berkeley, Berkeley, California 94720, USA
| | | | - Michael J Evans
- Department of Radiology, University of California at San Francisco School of Medicine, San Francisco, California 94107, USA
| | - Valerie B O'Donnell
- Systems Immunity Research Institute, Cardiff University, Cardiff CF14 4XN, United Kingdom
| | - Daniel K Nomura
- University of California at Berkeley, Berkeley, California 94720, USA
| | - Justin M Drake
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08903, USA
| | - Anna Ritz
- Department of Biology, Reed College, Portland, Oregon 97202, USA
| | - George V Thomas
- Knight Comprehensive Cancer Institute, Oregon Health and Science University, Portland, Oregon 97239, USA.,Department of Pathology and Laboratory Medicine, Oregon Health and Science University, Portland, Oregon 97239, USA
| |
Collapse
|
12
|
Mughal TI, Abdel-Wahab O, Rampal R, Mesa R, Koschmieder S, Levine R, Hehlmann R, Saglio G, Barbui T, Van Etten RA. Contemporary insights into the pathogenesis and treatment of chronic myeloproliferative neoplasms. Leuk Lymphoma 2016; 57:1517-26. [PMID: 27240645 PMCID: PMC6077976 DOI: 10.1080/10428194.2016.1185783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
This review is based on the deliberations at the 5th John Goldman Colloquium held in Estoril on 2nd October 2015 and the 9th post-ASH International Workshop on chronic myeloid leukemia (CML) and BCR-ABL1-negative myeloproliferative neoplasms (MPN) which took place on the 10th-11th December 2014, immediately following the 56th American Society of Hematology Annual Meeting. It has been updated since and summarizes the most recent advances in the biology and therapy of these diseases, in particular updates of genetics of MPN, novel insights from mouse MPN models, targeting CML stem cells and its niche; clinical advances include updates on JAK2 inhibitors and other therapeutic approaches to BCR-ABL1-negative MPNs, the use of alpha interferons, updates on tyrosine kinase inhibitors (TKI) randomized trials in CML, TKI cessation studies, and optimal monitoring strategies.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Disease Models, Animal
- Genetic Predisposition to Disease
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Mice
- Molecular Targeted Therapy
- Mutation
- Myeloproliferative Disorders/diagnosis
- Myeloproliferative Disorders/etiology
- Myeloproliferative Disorders/mortality
- Myeloproliferative Disorders/therapy
- Protein Kinase Inhibitors/administration & dosage
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/therapeutic use
- Risk Assessment
- Treatment Outcome
Collapse
Affiliation(s)
| | | | - Raajit Rampal
- b Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | - Ruben Mesa
- c Mayo Clinic Cancer Center , Scottsdale , AZ , USA
| | - Steffen Koschmieder
- d Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine , RWTH Aachen University , Aachen , Germany
| | - Ross Levine
- b Memorial Sloan Kettering Cancer Center , New York , NY , USA
| | | | | | - Tiziano Barbui
- g Papa Giovani XXIII Hospital and Research Center , Bergamo , Italy
| | - Richard A Van Etten
- h Chao Family Comprehensive Cancer Center, University of California Irvine , Irvine , CA , USA
| |
Collapse
|