119 results on '"Tremblay, Douglas"'
Search Results
2. Impact of Facility Type on Survival in Chronic Myelomonocytic Leukemia: A Propensity Score Matched, National Cancer Database Analysis.
- Author
-
Tharakan S, Feld J, Van Hyfte G, Mascarenhas J, and Tremblay D
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Aged, 80 and over, United States epidemiology, Adult, Survival Analysis, Leukemia, Myelomonocytic, Chronic mortality, Leukemia, Myelomonocytic, Chronic therapy, Propensity Score, Databases, Factual
- Abstract
Background: Chronic myelomonocytic leukemia (CMML) is a rare and likely underdiagnosed hematologic malignancy. Due to its rarity and nuances in diagnosis, many patients are referred to tertiary referral centers, although many continue to be cared for in the community setting. Given discrepancies in outcomes based on facility type in related myeloid malignancies, we hypothesized that CMML patients treated at academic centers may have improved survival as compared to patients treated at nonacademic centers (NACs)., Patients and Methods: Using the National Cancer Database (NCDB), we identified 6290 patients with CMML and collected data on demographics, comorbidities, treatment, and survival. We also performed a propensity matched analysis to control for baseline differences., Results: We found that patients at academic centers had higher median overall survival (OS) (17.7 months vs 14.7 months) and 5-year OS (19.1% vs 15.3%) than patients at NACs. In addition, patients treated at an academic center were also more likely to receive hematopoietic stem cell transplant as compared to those treated at NACs. Time to treatment initiation was overall similar between academic and NACs., Conclusion: Our study of one of the largest available datasets of CMML patients supports the importance of referring CMML patients to academic centers upon diagnosis to optimize outcomes in this rare hematologic malignancy., Competing Interests: Disclosure All authors have completed the ICMJE uniform disclosure form. J.M. receives clinical research funding paid to his institution from Incyte, Novartis, Celgene/BMS, CTI Bio, Geron, AbbVie, PharmaEssentia, Kartos, and Karyopharm. He receives consulting fees from Incyte, CTI Bio, Novartis, Geron, Kartos, Karyopharm, MorphoSys, AbbVie, Celgene/BMS, PharmaEssentia, Pfizer, Imago, Merck, Roche, GSK, Galecto. D.T. receives clinical research funding paid to his institution from Astellas Pharma, Gilead, CTI Bio and consulting fees from GSH, CTI Bio, Novartis, AbbVie, Sierra Oncology, and Cogent Biosciences. J.F. receives clinical research funding from Oryzon and Syros Pharmaceuticals. S.T. has no disclosures to report., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Evaluating the role of Day 14 bone marrow biopsy and European LeukemiaNet risk classification in predicting overall and relapse-free survival in acute myeloid leukemia.
- Author
-
Balev M, Zibara V, Van Hyfte G, Feld J, Kremyanskaya M, Becker M, Keyzner A, Shih AH, Marcellino B, Levavi H, Silverman L, Mascarenhas J, and Tremblay D
- Subjects
- Humans, Female, Male, Middle Aged, Disease-Free Survival, Aged, Adult, Bone Marrow pathology, Biopsy, Adolescent, Risk Assessment, Aged, 80 and over, Prognosis, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology
- Abstract
Multivariable analysis for overall survival and relapse-free survival demonstrating lack of significant for D14 BM status., (© 2024 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
4. Management of Advanced Systemic Mastocytosis: Clinical Challenges.
- Author
-
Tremblay D, Wagner NE, and Mascarenhas J
- Abstract
Advanced systemic mastocytosis (AdvSM) is a rare hematologic malignancy with organ damage and compromised life expectancy arising from organ accumulation of neoplastic mast cells. Identification of the gain-of-function KIT D816V in the majority of cases has accelerated pharmaceutical development culminating with the development of selective KIT inhibitors such as avapritinib. While the advent of these therapies has improved the quality and quantity of life in patients with AdvSM, current challenges remain in the management of this disease. In this review, we summarize the present and future therapeutics landscape of AdvSM, highlighting the development of novel KIT inhibitors including elenestinib and bezuclastinib. We also explore the continued role of additional treatment modalities including allogeneic stem cell transplantation before discussing unresolved clinical challenges in the management of AdvSM., Competing Interests: DT received research funding from Sobi, Sumitomo, Cogent Biosciences and Gilead and honoraria from Sobi, Novartis, AbbVie, PharmaEssentia, Sierra Oncology, GSK and Cogent Biosciences. JM received research funding from Incyte, BMS, Novartis, AbbVie, Geron, Kartos, Karyopharm, CTI/Sobi and PharmaEssentia and honoraria from Incyte, Blueprint Medicines, Keros, DISC Medicines, BMS, AbbVie, Kartos, Geron, GSK, Roche, Merck, Pfizer, PharmaEssentia, MorphoSys, Novartis, Galecto, Sobi, Sumitomo, and Karyopharm. The authors report no other conflicts of interest in this work., (© 2024 Tremblay et al.)
- Published
- 2024
- Full Text
- View/download PDF
5. Oral Decitabine/Cedazuridine Is an Effective Ambulatory Therapy for Patients With Myelofibrosis Refractory to JAK2 Inhibitor Therapy.
- Author
-
Handa S, Sivakumar G, Srisuwananukorn A, Dueck A, Tremblay D, Mascarenhas JO, Ginzburg Y, Kremyanskaya M, and Hoffman R
- Subjects
- Humans, Aged, Male, Female, Aged, 80 and over, Middle Aged, Retrospective Studies, Uridine analogs & derivatives, Uridine therapeutic use, Uridine pharmacology, Uridine administration & dosage, Administration, Oral, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Primary Myelofibrosis drug therapy, Primary Myelofibrosis mortality, Decitabine therapeutic use, Decitabine pharmacology, Decitabine administration & dosage, Janus Kinase 2 antagonists & inhibitors, Janus Kinase 2 genetics
- Abstract
Background: Outcomes are dismal for patients with myelofibrosis (MF) who are no longer responsive to JAK2 inhibitors (JAKi) and/or have increasing blast cell numbers. Although prior reports have suggested the benefits of intravenous decitabine (DAC) combined with ruxolitinib for patients with Myeloproliferative Neoplasm (MPN) accelerated/blast phase (AP/BP), decitabine-cedazuridine (DEC-C), an oral fixed-dose combination providing equivalent pharmacokinetic exposure, has not been evaluated in MF., Methods: We conducted a retrospective analysis of 14 patients with high-risk MF refractory to ruxolitinib or MPN-AP (10-19% blasts) treated with DEC-C +/- JAKi at Mount Sinai Hospital from 2021 to 2024., Results: The cohort was elderly (median age,76 years) and almost uniformly possessed high risk mutations with 13 of the 14 patients progressing on JAKi therapy. With a median follow-up of 9.4 months, the median overall survival (OS) was 29 months for the entire cohort. Median OS was 10.8 months for MPN-AP and was not reached for ruxolitinib refractory MF patients. All patients (n = 9) receiving > 4 cycles of DEC-C had clinical benefit exemplified by a reduction in blast cell numbers, spleen size, and lack of progression to MPN-BP (78%). Furthermore, 3/14 patients proceeded to allogeneic stem cell transplant. Myelosuppression was a common adverse event which was managed by reducing the number of days of administration of DEC-C from 5 to 3 per cycle., Conclusions: This report demonstrates the feasibility, tolerability, and clinical benefit of an exclusively ambulatory regimen for high-risk, elderly patients with advanced MF which warrants further evaluation in a prospective clinical trial., Competing Interests: Disclosures D.T. receives contracted research funding paid to his institution from Sobi, Astellas Pharma, Gilead and Cogent Biosciences and consulting fees from Novartis, AbbVie, Sierra Oncology, GSK and Cogent Biosciences. J.M. receives research funding paid to his institution from Incyte, Novartis, BMS, CTI/Sobi, Abbvie, Geron, Kartos and PharmaEssentia. Consulting fees paid to me by Incyte, Novartis, CTI/Sobi, Abbvie, Karyopharm, Kartos, Geron, GSK, Sumitomo, BMS, MorphoSys, Galecto, Pfizer, Merck, Roche and PharmaEssentia. R.H. received research grant funding through his institution from the following sources: Citi BioPharma Corp, Curis, Inc., Dexcel Pharma Technologies Ltd. Genentech, Kartos Therapeutics, Karyopharm Therapeutics, Inc., Kymera Therapeutics, OncoMyx Therapeutics, Novartis, Protagonist Therapeutics, Repare Therapeutics, Translational Drug Development (TD2), Turning Point Therapeutics, and the NIH/NCI. Additionally, he has received funding as an advisor/consultant from the following sources: AbbVie, Ionis Pharmaceuticals, Protagonist Therapeutics, Silence Therapeutics, GlycoMimetics, Dexcel Pharma Technologies LTD, Sumitomo Pharma Oncology, and Cellenkos, Inc. Finally, he holds a share of a patent filed by Dompé farmaceutici S.p.A. Y.Z.G. received research grant funding through her institution from the following sources: Protagonist Therapeutics, Repare Therapeutics, and the NIH/NIDDK. YZG has received funding as an advisor/consultant from the following sources: Bay Clinical, Takeda, Denali, Ionis Pharmaceuticals, Protagonist Therapeutics. No other relevant financial disclosures for other authors., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
6. Ven the dose matters: Venetoclax dosing in the frontline treatment of AML.
- Author
-
Sastow D, Levavi H, Wagner N, Pratz K, and Tremblay D
- Abstract
Older/unfit adults with AML have worse outcomes and fewer treatment options than their younger/fit counterparts. In vitro studies have found a synergistic effect of hypomethylating agents (HMA) with venetoclax (VEN) on AML cells and since the phase 3 VIALE-A trial demonstrated a survival benefit, HMA + VEN has become the standard of care in the frontline setting for older/unfit adults with AML. Unfortunately, the standard 28-day cycle of VEN is associated with a high degree of myelosuppression leading to treatment delays and dose modifications. Many small retrospective studies have successfully shown comparable outcomes to VIALE-A with reduced dose/duration of VEN. Furthermore, low dose metronomic dosing of HMA + VEN has shown clinical benefit while minimizing myelotoxicity. Future trials are vital to understand the appropriate dose of VEN in combination with HMA, to evaluate HMA + VEN compared to intensive therapy for younger/fit patients, and to explore its utility in the relapsed/refractory setting., Competing Interests: Declaration of competing interest Hannah Levavi receives consulting fees from Sobi. Keith Pratz receives research funding from AbbVie, Agios, Daiichi Sankyo, Millennium; advisory board member for AbbVie, Astellas, AstraZeneca, Boston Biomedical, Bristol Myers Squibb, Celgene, Novartis, Roche, Jazz Pharmaceuticals, and Servier. Douglas Tremblay receives contracted research funding paid to his institution from Sobi, Sumitomo, Cogent Biosciences and Gilead and consulting fees from Sobi, Novartis, AbbVie, Pharmaessentia, Sierra Oncology, GSK and Cogent Biosciences. All other authors have no conflicts of interest to disclose., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. How to customize common data models for rare diseases: an OMOP-based implementation and lessons learned.
- Author
-
Ahmadi N, Zoch M, Guengoeze O, Facchinello C, Mondorf A, Stratmann K, Musleh K, Erasmus HP, Tchertov J, Gebler R, Schaaf J, Frischen LS, Nasirian A, Dai J, Henke E, Tremblay D, Srisuwananukorn A, Bornhäuser M, Röllig C, Eckardt JN, Middeke JM, Wolfien M, and Sedlmayr M
- Subjects
- Humans, Rare Diseases
- Abstract
Background: Given the geographical sparsity of Rare Diseases (RDs), assembling a cohort is often a challenging task. Common data models (CDM) can harmonize disparate sources of data that can be the basis of decision support systems and artificial intelligence-based studies, leading to new insights in the field. This work is sought to support the design of large-scale multi-center studies for rare diseases., Methods: In an interdisciplinary group, we derived a list of elements of RDs in three medical domains (endocrinology, gastroenterology, and pneumonology) according to specialist knowledge and clinical guidelines in an iterative process. We then defined a RDs data structure that matched all our data elements and built Extract, Transform, Load (ETL) processes to transfer the structure to a joint CDM. To ensure interoperability of our developed CDM and its subsequent usage for further RDs domains, we ultimately mapped it to Observational Medical Outcomes Partnership (OMOP) CDM. We then included a fourth domain, hematology, as a proof-of-concept and mapped an acute myeloid leukemia (AML) dataset to the developed CDM., Results: We have developed an OMOP-based rare diseases common data model (RD-CDM) using data elements from the three domains (endocrinology, gastroenterology, and pneumonology) and tested the CDM using data from the hematology domain. The total study cohort included 61,697 patients. After aligning our modules with those of Medical Informatics Initiative (MII) Core Dataset (CDS) modules, we leveraged its ETL process. This facilitated the seamless transfer of demographic information, diagnoses, procedures, laboratory results, and medication modules from our RD-CDM to the OMOP. For the phenotypes and genotypes, we developed a second ETL process. We finally derived lessons learned for customizing our RD-CDM for different RDs., Discussion: This work can serve as a blueprint for other domains as its modularized structure could be extended towards novel data types. An interdisciplinary group of stakeholders that are actively supporting the project's progress is necessary to reach a comprehensive CDM., Conclusion: The customized data structure related to our RD-CDM can be used to perform multi-center studies to test data-driven hypotheses on a larger scale and take advantage of the analytical tools offered by the OHDSI community., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
8. Trends in location of death for individuals with acute myeloid leukemia in the United States.
- Author
-
Jain U, Rahim F, Jain B, Tresa Mathew A, Feliciano EJG, Dee EC, Lapen K, Lee F, Chino F, Tremblay D, and Tsai JJ
- Published
- 2024
- Full Text
- View/download PDF
9. Equivalent thrombotic risk with Warfarin, Dabigatran, or Enoxaparin after failure of initial direct oral anticoagulation (DOAC) therapy.
- Author
-
Shyu M, Liu A, Srikureja A, Gregorian A, Srisuwananukorn A, Tremblay D, and Naymagon L
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Administration, Oral, Hemorrhage chemically induced, Retrospective Studies, Treatment Failure, Thrombosis prevention & control, Thrombosis chemically induced, Thrombosis etiology, Thrombosis drug therapy, Rivaroxaban adverse effects, Rivaroxaban administration & dosage, Rivaroxaban therapeutic use, Pyrazoles, Pyridones, Dabigatran adverse effects, Dabigatran administration & dosage, Dabigatran therapeutic use, Enoxaparin adverse effects, Enoxaparin administration & dosage, Anticoagulants adverse effects, Anticoagulants administration & dosage, Anticoagulants therapeutic use, Warfarin adverse effects, Warfarin administration & dosage, Venous Thromboembolism drug therapy
- Abstract
Background: The direct oral anticoagulants (DOACs) are now commonly regarded as first line anticoagulants in most cases of venous thromboembolism (VTE). However, the optimal choice of subsequent anticoagulant in instances of first line DOAC failure is unclear., Objectives: To describe and compare outcomes with second line anticoagulants used after DOAC failure., Methods: Patients seen at an urban hospital system for an episode of acute VTE initially treated with either apixaban or rivaroxaban who experienced a subsequent recurrent thrombosis while on anticoagulation (1st recurrent thrombosis) were included., Results: In total, 166 patients after apixaban or rivaroxaban failure were included. Following DOAC failure (1st recurrent thrombosis), the subsequent anticoagulant was warfarin in 60 patients (36%), dabigatran in 42 patients (25%), and enoxaparin in 64 patients (39%). Enoxaparin was preferentially prescribed in patients with a malignancy-associated etiology for 1st recurrent thrombosis (p < 0.01). The median follow-up time in our cohort was 16 months. There was no difference in 2nd recurrent thrombosis-free survival (p = 0.72) or risk for major bleeding event (p = 0.30) among patients treated with dabigatran, warfarin, or enoxaparin., Conclusions: In this retrospective analysis of patients failing first line DOAC therapy, rates of 2nd recurrent thrombosis and bleeding did not differ among subsequently chosen anticoagulants. Our study provides evidence that the optimal 2nd anticoagulant is not clear, and the choice of 2nd anticoagulant should continue to balance patient preference, cost, and provider experience., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
10. Conventional Cytogenetic Analysis and Array CGH + SNP Identify Essential Thrombocythemia and Prefibrotic Primary Myelofibrosis Patients Who Are at Risk for Disease Progression.
- Author
-
Tripodi J, Hoffman R, Tremblay D, Ahire D, Mascarenhas J, Kremyanskaya M, and Najfeld V
- Subjects
- Humans, Comparative Genomic Hybridization, Polymorphism, Single Nucleotide, Retrospective Studies, Cytogenetic Analysis, Disease Progression, Thrombocythemia, Essential diagnosis, Thrombocythemia, Essential genetics, Primary Myelofibrosis diagnosis, Primary Myelofibrosis genetics
- Abstract
The Philadelphia chromosome-negative myeloproliferative neoplasms (Ph-MPNs) are a heterogeneous group of clonal hematopoietic malignancies that include polycythemia vera (PV), essential thrombocythemia (ET), and the prefibrotic form of primary myelofibrosis (prePMF). In this study, we retrospectively reviewed the karyotypes from conventional cytogenetics (CC) and array Comparative Genomic Hybridization + Single Nucleotide Polymorphism (aCGH + SNP) in patients with ET or prePMF to determine whether the combined analysis of both methodologies can identify patients who may be at a higher risk of disease progression. We performed a comprehensive genomic review on 169 patients with a clinical diagnosis of ET (154 patients) or prePMF (15 patients). Genomic alterations detected by CC or array-CGH + SNP were detected in 36% of patients. In patients who progressed, 68% had an abnormal genomic finding by either technology. There was a shorter progression-free survival (PFS) among patients who were cytogenetically abnormal or who were cytogenetically normal but had an abnormal aCGH + SNP result. Leveraging the ability to detect submicroscopic copy number alterations and regions of copy neutral-loss of heterozygosity, we identified a higher number of patients harboring genomic abnormalities than previously reported. These results underscore the importance of genomic analysis in prognostication and provide valuable information for clinical management and treatment decisions.
- Published
- 2024
- Full Text
- View/download PDF
11. Systematic review and meta-analysis evaluating clinical outcomes in adult acute myeloid leukemia patients with central nervous system involvement.
- Author
-
Goulart H, Sastow D, Moshier E, Martin L, Mascarenhas J, and Tremblay D
- Subjects
- Adult, Humans, Prognosis, Survival Rate, Treatment Outcome, Central Nervous System Neoplasms mortality, Central Nervous System Neoplasms therapy, Central Nervous System Neoplasms pathology, Central Nervous System Neoplasms diagnosis, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute diagnosis
- Abstract
Patients with acute myeloid leukemia (AML) may experience extramedullary involvement when disease is present outside of the blood and bone marrow. In particular, the presence of central nervous system (CNS) involvement has traditionally been thought of as a poor prognostic factor. In the presently available literature, there is a paucity of conclusive data surrounding CNS AML given its rarity and lack of unified screening practices. Thus, we performed a systematic review and meta-analysis in order to more definitively characterize survival outcomes in this patient population. In this meta-analysis, we evaluated survival outcomes and response rates from clinical studies on patients with AML stratified by the presence of CNS involvement. Twelve studies were included in the meta-analysis with a resulting hazard ratio (HR) for overall survival (OS) of 1.34 with a 95 % CI of 1.14 to 1.58. These findings suggest that CNS involvement in adult patients with AML is associated with an increased hazard of mortality compared to those patients without CNS involvement. As such, CNS involvement should be viewed as negative prognostic marker, and attention should be made to ensure prompt identification and treatment of patients who experience this complication., Competing Interests: Declaration of Competing Interest JM has received research funding paid to the institution from Novartis, CTI, Incyte, BMS, Geron, Abbvie, Kartos and PharmaEssentia and consulting fees from Incyte, CTI Bio, Novartis, Roche, GSK, Sierra Onc, PharmaEssentia, Kartos, Karyopharm, Galecto, Imago, BMS, Abbvie and Geron. DT has received research funding paid to his institution from CTI Biopharma, Astellas Pharma and Gilead and consulting fees from CTI Biopharma, Novartis, AbbVie, Sierra Oncology, GSK and Cogent. The other authors have no competing interests to report., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Understanding triple negative myeloproliferative neoplasms: pathogenesis, clinical features, and management.
- Author
-
Tharakan S, Mascarenhas J, and Tremblay D
- Subjects
- Humans, Mutation, Janus Kinase 2 genetics, Calreticulin genetics, Myeloproliferative Disorders genetics, Primary Myelofibrosis genetics, Thrombocythemia, Essential genetics, Neoplasms
- Abstract
ABSTRACT Myeloproliferative neoplasms (MPNs) that lack the classical "driver mutations," termed triple negative MPNs, remain a poorly understood entity. Despite considerable progress toward understanding MPN pathobiology, the mechanisms leading to the development of these MPNs remains inadequately elucidated. While triple negative primary myelofibrosis (TN-PMF) portends a poor prognosis, triple negative essential thrombocythemia (TN-ET) is more favorable as compared with JAK2 mutated ET. In this review, we summarize the clinical features and prognosis of TN-PMF and -ET as well as diagnostic challenges including identification of non-canonical driver mutations. We also discuss additional molecular drivers to better understand possible pathogenic mechanisms underlying triple negative MPNs. Finally, we highlight current therapeutic approaches as well as novel targets, particularly in the difficult to treat TN-PMF population.
- Published
- 2024
- Full Text
- View/download PDF
13. Decreased patency of transjugular intrahepatic portosystemic shunts performed for splanchnic vein thrombosis in patients with myeloproliferative neoplasms.
- Author
-
Shyu M, Winters A, Naymagon L, Patel R, Schiano TD, and Tremblay D
- Subjects
- Humans, Portal Vein, Portasystemic Shunt, Transjugular Intrahepatic adverse effects, Neoplasms, Venous Thrombosis complications, Venous Thrombosis surgery, Myeloproliferative Disorders complications, Myeloproliferative Disorders surgery
- Abstract
Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Douglas Tremblay receives contracted research funding paid to his institution from CTI Biopharma, Astellas Pharma and Gilead and consulting fees from CTI Biopharma, Novartis, AbbVie, Sierra Oncology, GSK and Cogent Biosciences. There are no other conflicts of interest to disclose.
- Published
- 2024
- Full Text
- View/download PDF
14. Response to 'blast identification in cerebrospinal fluid specimens from patients with acute myeloid leukemia: laboratory perspectives'.
- Author
-
Sastow D, Tatarian J, Salib C, Lin TL, and Tremblay D
- Subjects
- Humans, Blast Crisis, Leukemia, Myeloid, Acute diagnosis
- Published
- 2024
- Full Text
- View/download PDF
15. Exclusion of Acute Myeloid Leukemia Patients with Central Nervous System Involvement from Clinical Trials: An Analysis of the National Institutes of Health Clinical Trials Registry from 2012 to 2022.
- Author
-
Sastow D, Van Hyfte G, Feld J, Kremyanskaya M, Mascarenhas J, and Tremblay D
- Subjects
- Humans, United States, National Institutes of Health (U.S.), Patient Selection, Adult, Female, Male, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute diagnosis, Registries, Central Nervous System Neoplasms therapy, Clinical Trials as Topic
- Abstract
Introduction: Central nervous system (CNS) involvement in acute myeloid leukemia (AML) can be successfully treated with intrathecal chemotherapy and carries debatable prognostic impact. However, patients with CNS involvement are commonly excluded from clinical trials at an unknown rate. We systematically evaluated exclusion criteria of AML clinical trials based on CNS involvement and determined associations with clinical trial characteristics., Methods: The National Institutes of Health Clinical Trials Registry was searched for interventional adult AML trials between 2012 and 2022 that were phase 1, 2, or 3 and relevant trial characteristics were extracted., Results: 1,270 trials were included in the analysis with 790 trials (62.1%) explicitly excluding CNS involvement. There was no significant change in rates of CNS exclusion over the past decade. CNS exclusion was higher in trials that included the non-transplant population compared to trials exclusive to the transplant population (66.9% vs. 43.8%, p < 0.01). Non-transplant trials were also more likely to exclude patients with a history of or ambiguous timing of CNS involvement (p < 0.01). Phase 3 trials were associated with more liberal definitions of CNS exclusion (history or ambiguous timing) as compared to phase 1 and 2 trials that had higher rates of excluding patients with only active CNS involvement (p < 0.01)., Conclusion: A majority of AML clinical trials, particularly in the non-transplant setting, exclude patients with CNS involvement. Many of these trials, most notably phase 3 trials, exclude patients not only with active but also with any history of CNS involvement. Further research is needed to determine optimal management of these patients in order to increase representation in large clinical trials., (© 2023 S. Karger AG, Basel.)
- Published
- 2024
- Full Text
- View/download PDF
16. Discontinuation Syndrome With JAK2 Selective Agents: Case Presentation and Mechanistic Insights.
- Author
-
Handa S, Farina KA, Becker M, Kelly B, Yu A, Feld J, Tremblay D, Marcellino BK, Salib C, Mascarenhas J, and Shih AH
- Subjects
- Humans, Bridged-Ring Compounds, Pyrimidines, Janus Kinase 2 genetics, Primary Myelofibrosis
- Abstract
We report the first case of pacritinib-withdrawal syndrome with in vitro elucidation of underlying mechanisms.
- Published
- 2024
- Full Text
- View/download PDF
17. Cytoreduction for ET and PV: who, what, when, and how?
- Author
-
Tremblay D
- Subjects
- Humans, Cytoreduction Surgical Procedures adverse effects, Hydroxyurea, Polycythemia Vera diagnosis, Thrombocythemia, Essential diagnosis, Thrombosis etiology
- Abstract
Thrombotic complications are the primary contributor to morbidity and mortality in essential thrombocythemia (ET) and polycythemia vera (PV). Cytoreductive therapy is the main tool for primary or tertiary thrombosis prevention in these diseases. In general, high-thrombotic-risk patients and those with symptoms that may be ameliorated from cytoreductive therapy are candidates for this treatment, although the decision is highly individualized. Approved options for cytoreduction in ET and PV include hydroxyurea, long-acting interferons, anagrelide in ET, and ruxolitinib in PV. Selecting the ideal agent requires careful consideration of the toxicity profiles and individual treatment goals. In this review the existing literature on cytoreductive decisions in ET and PV is summarized, with an emphasis on risk-stratification, highlighting the need for personalized care in order to maximize the benefit of these therapies while minimizing toxicities., (Copyright © 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
18. Multi-institutional analysis of outcomes in acute myeloid leukemia patients with central nervous system involvement.
- Author
-
Sastow D, Tatarian J, Riazat-Kesh YJRA, Farina K, Becker M, Feld J, Kremyanskaya M, Mascarenhas J, Byrd K, Male HJ, Lin TL, and Tremblay D
- Subjects
- Adult, Humans, Recurrence, Central Nervous System, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Central Nervous System Neoplasms diagnosis, Central Nervous System Neoplasms drug therapy
- Abstract
We evaluated outcomes of AML patients with central nervous system (CNS) involvement at two academic institutions. Fifty-two adult patients were identified. Neurologic symptoms were reported in 69% of patients, with headache the most common (33%). 84% ( n = 42) of patients cleared their cerebrospinal fluid (CSF), with a median number of one dose of intrathecal (IT) chemotherapy. Of these patients, 21% ( n = 9) had a CSF relapse, with 67% ( n = 6) of those experiencing CSF relapse also having concurrent bone marrow relapse. Of the 36 patients with baseline neurologic symptoms, 69% had improvement in symptoms post-IT therapy. The median overall survival was 9.3 months and 3.5 months for patients with CNS involvement diagnosed before/during induction and at relapse, respectively. In this study, IT therapy was rapidly effective in clearing CSF blasts and improving neurologic symptoms in most patients. Few patients experienced CSF relapse, which predominantly occurred in the setting of concomitant bone marrow relapse.
- Published
- 2023
- Full Text
- View/download PDF
19. An overlooked mimic? Autoimmune myelofibrosis-A scoping review of the literature.
- Author
-
Amel Riazat-Kesh YJR, Maraveyas A, Martin L, and Tremblay D
- Subjects
- Humans, Male, Female, Adult, Retrospective Studies, Bone Marrow pathology, Immunosuppression Therapy, Primary Myelofibrosis diagnosis, Primary Myelofibrosis therapy, Primary Myelofibrosis complications, Autoimmune Diseases diagnosis, Autoimmune Diseases therapy, Autoimmune Diseases complications
- Abstract
Background and Objectives: Autoimmune myelofibrosis (AIMF) is a rare cause of bone marrow fibrosis (BMF) occurring in the presence or absence of a defined autoimmune disease (secondary or primary AIMF, sAIMF/pAIMF, respectively). Unlike primary myelofibrosis (PMF), AIMF responds well to immunosuppressive therapy with a benign clinical course. Diagnostic criteria for AIMF in opposition to PMF have been lacking, though recent work has helped better characterise molecular and pathological features of AIMF, improving diagnostic precision., Methods: Using a modern clinical and pathophysiological understanding of AIMF, we apply scoping review methodology and rigorous case-criteria to retrospectively analyse the case literature. We examine its patient-population, describing patient-associated factors, presentation, bone marrow pathology, genetics, treatment and outcomes., Results: Fifty-five studies were identified, describing 139 AIMF patients. Patients were mostly young females (~4:1 ratio female:male, median age 40.8 years) and typically presented with cytopenias. Splenomegaly was rare. sAIMF was more common than pAIMF (~3:1 ratio), and most cases responded well to immunosuppressive therapy., Conclusions: Our results strengthen the emerging picture of AIMF's patient population, natural history and response to treatment. Further work should continue to use reproducible diagnostic criteria, and explore AIMF's pathophysiology, response to different therapies, and sequelae over larger timescales, as well as differences between pAIMF, sAIMF and PMF., (© 2023 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
20. The core concepts of core binding factor acute myeloid leukemia: Current considerations for prognosis and treatment.
- Author
-
Darwish C, Farina K, and Tremblay D
- Subjects
- Humans, Prognosis, Cytarabine therapeutic use, Core Binding Factors genetics, Recurrence, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute etiology, Leukemia, Myeloid, Acute therapy
- Abstract
Core binding factor acute myeloid leukemia (CBF AML), defined by t(8;21) or inv(16), is a subset of favorable risk AML. Despite its association with a high complete remission rate after induction and relatively good prognosis overall compared with other subtypes of AML, relapse risk after induction chemotherapy remains high. Optimizing treatment planning to promote recurrence free survival and increase the likelihood of survival after relapse is imperative to improving outcomes. Recent areas of research have included evaluation of the role of gemtuzumab in induction and consolidation, the relative benefit of increased cycles of high dose cytarabine in consolidation, the utility of hypomethylating agents and kinase inhibitors, and the most appropriate timing of stem cell transplant. Surveillance with measurable residual disease testing is increasingly being utilized for monitoring disease in remission, and ongoing investigation seeks to determine how to use this tool for early identification of patients who would benefit from proceeding to transplant. In this review, we outline the current therapeutic approach from diagnosis to relapse while highlighting the active areas of investigation in each stage of treatment., Competing Interests: Conflict of interest Douglas Tremblay receives contracted research funding paid to his institution from CTI Biopharma, Astellas Pharma, and Gilead and consulting fees from CTI Biopharma, Novartis, AbbVie, Sierra Oncology, GSK, and Cogent. The rest of the authors have no conflicts of interest to disclose., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. Dasatinib-Associated Acneiform Eruption Successfully Treated With Sarecycline in a Patient With Chronic Myeloid Leukemia.
- Author
-
Poplausky D, Young JN, Dubin DP, Tremblay D, and Gulati N
- Abstract
Dasatinib is a second-generation tyrosine inhibitor that is used for the treatment of patients with chronic myeloid leukemia (CML). It can cause a myriad of skin toxicities, including pruritis, pigmentary abnormalities of hair and skin, and maculopapular rashes. Rarely, it can be associated with acneiform eruptions, which are typically treated with doxycycline. However, doxycycline may not be an ideal therapy, especially for long-term use, due to the risk of gut flora disruption, antimicrobial resistance, and side effects. We present a case of a CML patient who developed an acneiform eruption associated with dasatinib and was successfully treated with sarecycline, a narrow-spectrum tetracycline. Given its targeted spectrum of activity, sarecycline has a lower risk of antimicrobial resistance and an improved safety profile compared to first- and second-generation tetracyclines such as doxycycline. As acneiform drug eruptions can have a significant impact on a patient's quality of life, effective management by dermatologists is paramount. Sarecycline may be a suitable treatment with a favorable safety profile, making it an appropriate choice for patients, especially those who require long-term therapy., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2023, Poplausky et al.)
- Published
- 2023
- Full Text
- View/download PDF
22. CNL and aCML are prognostically distinct: a large National Cancer Database analysis.
- Author
-
Tremblay D, Sastow D, and Mascarenhas J
- Subjects
- Humans, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative, Neoplasms diagnosis, Neoplasms epidemiology
- Published
- 2023
- Full Text
- View/download PDF
23. Energy expenditure in myelofibrosis patients treated with a JAK1/2 inhibitor.
- Author
-
Tremblay D, Dougherty M, Mascarenhas J, and Gallagher EJ
- Subjects
- Humans, Pilot Projects, Nitriles, Weight Gain, Energy Metabolism, Janus Kinase 1, Primary Myelofibrosis drug therapy, Primary Myelofibrosis complications
- Abstract
Weight gain is a known adverse effect of ruxolitinib, a JAK1/2 inhibitor that is the mainstay of treatment for many patients with myelofibrosis. The mechanisms behind weight increase with ruxolitinib is incompletely understood, although decreased adipose tissue lipolysis and increased appetite due to blocking the effects of leptin in the hypothalamus have been proposed. In order to explore the metabolic changes in ruxolitinib-treated patients with myelofibrosis, we performed a pilot study to assess the feasibility of using a portable indirect calorimeter to quantify energy expenditure before and during ruxolitinib treatment and report the results of two patients. Waist circumference increased during ruxolitinib treatment in both patients. Energy expenditure initially increased followed by a decrease and then increase again, but to levels below baseline. These results suggest that weight gain secondary to ruxolitinib may be related to changes in whole body energy expenditure., Competing Interests: DT receives research funding paid to his institution from CTI Biopharma, Astellas Pharma and Gilead and consulting fees from AbbVie, Sierra Oncology, GSK and Cogent. JM receives research funding paid to his institution from Incyte, CTI Bio, Novartis, PharmaEssentia, Merck, Kartos, Geron, BMS, Roche, and consulting fees from Incyte, CTI Bio, Novartis, Roche, BMS, Celgene, Geron, PharmaEssentia, Morphosys, Sierra Oncology, GSK, Galecto, Karyopharm, Kartos, and Imago. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Tremblay, Dougherty, Mascarenhas and Gallagher.)
- Published
- 2023
- Full Text
- View/download PDF
24. Emerging Treatment Options for Myelofibrosis: Focus on Anemia.
- Author
-
Sastow D and Tremblay D
- Abstract
Myelofibrosis (MF) is a hematologic malignancy characterized by abnormal proliferation of myeloid cells and the release of pro-inflammatory cytokines, leading to progressive bone marrow dysfunction. The introduction of ruxolitinib just over a decade ago marked a significant advancement in MF therapy, with JAK inhibitors now being the first-line treatment for reducing spleen size and managing symptoms. However, early JAK inhibitors (ruxolitinib and fedratinib) are often associated with cytopenias, particularly thrombocytopenia and anemia, which limit their tolerability. To address these complications, pacritinib has been developed and recently approved for patients with thrombocytopenia, while momelotinib is in development for those with anemia. Although JAK inhibitors have significantly improved the quality of life of MF patients, they have not demonstrated the ability to reduce leukemic transformation and their impact on survival is debated. Numerous drugs are currently being developed and investigated in clinical trials, both as standalone therapy and in combination with JAK inhibitors, with promising results enhancing the benefits of JAK inhibitors. In the near future, MF treatment strategies will involve selecting the most suitable JAK inhibitor based on individual patient characteristics and prior therapy. Ongoing and future clinical trials are crucial for advancing the field and expanding therapeutic options for MF patients., Competing Interests: Dr Douglas Tremblay reports grants, personal fees from CTI Biopharma, grants from Astellas Pharma, grants from Gilead, personal fees from Novartis, personal fees from AbbVie, personal fees from Sierra Oncology, personal fees from GSK, personal fees from Cogent Biosciences, outside the submitted work. The authors report no other conflicts of interest in this work., (© 2023 Sastow and Tremblay.)
- Published
- 2023
- Full Text
- View/download PDF
25. Pharmacotherapeutic advances for splenomegaly in myelofibrosis.
- Author
-
Tremblay D and Mascarenhas J
- Subjects
- Humans, Bridged-Ring Compounds therapeutic use, Treatment Failure, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors pharmacology, Nitriles therapeutic use, Janus Kinase 2, Splenomegaly drug therapy, Splenomegaly etiology, Primary Myelofibrosis complications, Primary Myelofibrosis drug therapy
- Abstract
Introduction: Myelofibrosis is a hematologic malignancy with a variety of clinical manifestations including splenomegaly, which is present in approximately 80% of newly diagnosed patients. JAK inhibitors are the mainstay of pharmacologic treatment for splenomegaly in myelofibrosis, although spleen size reduction is not universal, and the duration of benefit is only moderately durable., Areas Covered: We first discuss the pathobiology of splenomegaly in myelofibrosis before detailing approved and novel pharmacotherapies that can reduce spleen size while also highlighting non-pharmacologic approaches. In this review, efficacy of these treatments is measured solely by spleen volume reduction, acknowledging that other outcome measures such as symptom improvement and survival are also critical., Expert Opinion: Currently, ruxolitinib can be administered to the majority of frontline patients although those with severe thrombocytopenia should receive pacritinib to address spleen burden. Momelotinib may be particularly well suited for patients with significant anemia and novel combination treatments in clinical development may improve the depth and duration of spleen responses. After frontline treatment failure, fedratinib, or pacritinib are commercial options for patients with persistent symptomatic splenomegaly. Novel agents given alone or in combination with a JAK inhibitor are being explored in trials, which may ameliorate splenomegaly and ultimately improve disease progression.
- Published
- 2023
- Full Text
- View/download PDF
26. Ascertaining QUAZARs: slow-motion and light-speed development of oral azacitidine and decitabine.
- Author
-
Feld J, Tremblay D, Navada SC, and Silverman LR
- Subjects
- Humans, Decitabine therapeutic use, Antimetabolites, Antineoplastic therapeutic use, Azacitidine therapeutic use, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes pathology, Leukemia, Myeloid, Acute drug therapy
- Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are devastating diseases that frequently rely on the use of parenteral hypomethylating agents (HMAs), either as monotherapy or in combination, as first-line treatment for many patients. Two new oral HMAs, decitabine/cedazuridine (DC) for use in place of azacitidine or decitabine in MDS, and azacitidine (CC-486) for use as maintenance treatment in AML, were recently approved by the FDA. We will discuss the development of these oral HMAs, including the advantages/disadvantages in transitioning to oral HMAs and an in depth look at the pivotal phase III trials that led to their FDA approval - ASCERTAIN for DC and QUAZAR-AML-001 for CC-486. We also review how these agents have been and are being studied in other malignancies, and examine the future role that these exciting novel agents will play in both MDS and AML.
- Published
- 2023
- Full Text
- View/download PDF
27. New Treatments for Myelofibrosis.
- Author
-
Tremblay D and Mesa R
- Subjects
- Humans, Janus Kinases metabolism, STAT Transcription Factors metabolism, Signal Transduction, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors pharmacology, Primary Myelofibrosis diagnosis, Primary Myelofibrosis drug therapy, Primary Myelofibrosis etiology, Janus Kinase Inhibitors therapeutic use
- Abstract
Opinion Statement: Currently approved therapies for myelofibrosis (MF) consist of JAK inhibitors, which produce meaningful improvements in spleen size and symptom burden but do not significantly impact leukemic progression. In addition, many patients develop resistance or intolerance to existing therapies and are left without meaningful therapeutic options. There has been recent rapid development of agents in MF that may be able to fill these unmet needs. Importantly, most treatments currently in clinical development have targets outside the JAK-STAT pathway, including BET, BCL-2/BCL-xL, PI3k, HDM2, PIM-1, SINE, telomerase, LSD1, and CD123. These therapies are being tested in combination with JAK inhibitors in the front-line setting and in patients with a suboptimal response, as well as a single agent after JAK inhibitor failure. This next generation of agents is likely to produce a paradigm shift in MF treatment with a focus on combination treatment targeting multiple areas of MF pathophysiology., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
28. Splanchnic Vein Thrombosis in Myeloproliferative Neoplasms: Treatment Considerations and Unmet Needs.
- Author
-
Liu A, Naymagon L, and Tremblay D
- Abstract
Patients who develop splanchnic vein thrombosis (SVT) in the setting of a myeloproliferative neoplasm (MPN) are at risk for complications including portal hypertension, bleeding, thrombosis, and death. Prompt multidisciplinary treatment is thus necessary to prevent long-term sequelae. However, optimal management strategies are not well established due to a paucity of data. In this review, we very briefly discuss the epidemiology, pathophysiology, and prognosis of MPN-SVT and then more comprehensively explore treatment considerations of MPN-SVT, including anticoagulation, endovascular/surgical intervention, and cytoreductive therapy. We will also highlight current gaps in our knowledge of MPN-SVT and conclude by suggesting future directions to optimize the treatment of MPN-SVT and improve outcomes.
- Published
- 2022
- Full Text
- View/download PDF
29. Thrombocytopenia in Patients With Myelofibrosis: A Practical Management Guide.
- Author
-
Tremblay D, Baine I, and Mascarenhas J
- Subjects
- Humans, Janus Kinase 2, Protein Kinase Inhibitors therapeutic use, Primary Myelofibrosis complications, Primary Myelofibrosis diagnosis, Primary Myelofibrosis therapy, Thrombocytopenia chemically induced, Anemia chemically induced
- Abstract
Patients with myelofibrosis (MF) frequently develop thrombocytopenia as a consequence of bone marrow fibrosis, splenic sequestration, and myelosuppression from an inflammatory microenvironmental milieu. Thrombocytopenia occurs frequently at diagnosis, worsens with disease progression, is an independent adverse prognostic factor, and limits effective dosing of JAK2 inhibitors. Recently, pacritinib was approved for patients with MF and extreme thrombocytopenia. However, this JAK2/IRAK1 inhibitor is not primarily used to attain improvement in platelet count. In this narrative review, we discuss strategies to specifically address thrombocytopenia in MF patients including immunomodulatory drugs, synthetic androgens, hypomethylating agents and splenectomy, all of which have only modest efficacy in alleviating thrombocytopenia. We also detail transfusion approaches, including diagnostic and therapeutic consideration for platelet transfusion refractoriness. We end by discussing novel therapies, including TGFβ traps and recombinant pentraxin-2, which may increase platelet counts in MF patients. Despite recent therapeutic advancements in MF, there remains a near paucity of agents that can effectively alleviate thrombocytopenia., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
30. Efficacy and Safety of Pacritinib vs Placebo for Patients With Severe COVID-19: A Phase 2 Randomized Clinical Trial.
- Author
-
Cafardi J, Miller C, Terebelo H, Tewell C, Benzaquen S, Park D, Egan P, Lebovic D, Pettit K, Whitman E, Tremblay D, Feld J, Buckley S, Roman-Torres K, Smith J, Craig A, and Mascarenhas J
- Subjects
- Adult, Humans, Male, Middle Aged, Interleukin-6, Pyrimidines, SARS-CoV-2, Female, Young Adult, Aged, Aged, 80 and over, COVID-19 Drug Treatment, Janus Kinase Inhibitors therapeutic use
- Abstract
Importance: The morbidity and mortality associated with COVID-19 remain high despite advances in standard of care therapy, and the role of anti-inflammatory agents that inhibit the interleukin 6/JAK2 pathway is still being elucidated., Objective: To evaluate the efficacy and safety of the oral JAK2/IRAK1 inhibitor pacritinib vs placebo in the treatment of adults with severe COVID-19., Design, Setting, and Participants: This phase 2, double-blind, placebo-controlled, randomized clinical trial enrolled hospitalized adult patients with severe COVID-19 at 21 centers across the US between June 2020 and February 2021, with approximately 1.5 months of safety follow-up per patient. Data analysis was performed from September 2021 to July 2022., Interventions: Patients were randomized 1:1 to standard of care plus pacritinib (400 mg per os on day 1 followed by 200 mg twice daily on days 2-14) vs placebo, for 14 days., Main Outcomes and Measures: The primary end point was death or need for invasive mechanical ventilation (IMV) or extracorporeal membrane oxygenation (ECMO) by day 28. All-cause mortality and safety were also assessed., Results: A total of 200 patients were randomized to pacritinib (99 patients; 56 men [56.6%]; median [range] age, 60 [19-87] years) or placebo (101 patients; 64 men [63.4%]; median [range] age 59 [28-94] years). The percentage requiring supplementary oxygen was 99.0% (98 patients) in the pacritinib group vs 98.0% (99 patients) in the placebo group. The percentage who progressed to IMV, ECMO, or death was 17.2% (17 patients) in the pacritinib group vs 22.8% (23 patients) in the placebo group (odds ratio, 0.62; 95% CI, 0.28-1.35; P = .23). Among patients with elevated interleukin 6, the rate was 17.5% (11 of 63 patients) in the pacritinib group vs 30.4% (21 of 96 patients) in the placebo group. The adverse event rate was similar for pacritinib vs placebo (78.1% [75 patients] vs 80.2% [81 patients]), with no excess in infection (14.6% [14 patients] vs 19.8% [20 patients]), bleeding (8.3% [8 patients] vs 10.9% [11 patients]), or thrombosis (8.3% [8 patients] vs 7.9% [8 patients]). Rates of grade 3 or higher adverse events were lower with pacritinib than placebo (29.2% [28 patients] vs 40.6% [41 patients])., Conclusions and Relevance: The study did not meet its primary end point in patients with severe COVID-19. Subgroup analyses may indicate specific populations with hyperinflammation that could benefit from pacritinib, although further clinical trials would be needed to confirm these effects., Trial Registration: ClinicalTrials.gov Identifier: NCT04404361.
- Published
- 2022
- Full Text
- View/download PDF
31. Impact of Fluoroquinolone Prophylaxis on Neutropenic Fever, Infections, and Antimicrobial Resistance in Newly Diagnosed AML Patients.
- Author
-
Caro J, Madero-Marroquin R, Zubizarreta N, Moshier E, Tremblay D, Coltoff A, Lancman G, Fuller R, Rana M, Mascarenhas J, and Jacobs SE
- Subjects
- Adult, Humans, Fluoroquinolones pharmacology, Fluoroquinolones therapeutic use, Anti-Bacterial Agents therapeutic use, Retrospective Studies, Prospective Studies, Drug Resistance, Bacterial, Clostridioides difficile, Bacterial Infections prevention & control, Leukemia, Myeloid, Acute complications, Leukemia, Myeloid, Acute drug therapy
- Abstract
Introduction: Fluoroquinolone prophylaxis is recommended during induction chemotherapy for patients with acute myeloid leukemia (AML) to reduce risk of neutropenic fever and systemic bacterial infections. We evaluated the effectiveness of primary fluoroquinolone prophylaxis in an area with high fluoroquinolone resistance., Materials and Methods: We performed a retrospective chart review of newly diagnosed adult AML patients who received frontline therapy at Mount Sinai Hospital in New York, NY, between 2012 and 2019. Primary outcome was development of neutropenic fever. Secondary outcomes were development of systemic bacterial infections and infections with multidrug-resistant organisms and Clostridioides difficile. Infectious outcomes were collected through 6 months after therapy initiation. We estimated the effect of fluoroquinolone prophylaxis with a time-dependent Cox proportional hazards model., Results: Of 121 included patients, 87 received antibiotic prophylaxis and 34 did not. There was no difference in baseline characteristics, although the prophylaxis group had longer neutropenia duration (median 30 vs. 23 days, P = .013). The prophylaxis group had a reduced risk of neutropenic fever (hazard ratio 0.59, P = .039). The prophylaxis group had fewer gram-positive (P = .043) and gram-negative (P = .049) bloodstream infections and fewer clinically documented infections during frontline therapy (P = .005) and follow-up (P = .026). There was no difference in incidence of C. difficile or infection with fluoroquinolone-resistant or multidrug-resistant organisms. There was no mortality difference between groups., Conclusion: In an area with high fluoroquinolone resistance, primary fluoroquinolone prophylaxis in newly diagnosed AML patients reduced the risk of neutropenic fever and systemic bacterial infections without increased antimicrobial resistance. Prospective, randomized studies are needed to confirm these observations., (Copyright © 2022. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
32. Splanchnic vein thrombosis associated with myeloproliferative neoplasms.
- Author
-
Tremblay D, Winters A, Beckman JD, Naymagon L, Patel R, Mascarenhas J, and Schiano TD
- Subjects
- Anticoagulants therapeutic use, Endothelial Cells pathology, Humans, Splanchnic Circulation, Myeloproliferative Disorders complications, Neoplasms complications, Venous Thrombosis pathology
- Abstract
Splanchnic vein thrombosis (SVT) in the setting of myeloproliferative neoplasm (MPN) is a unique clinical entity that requires close interdisciplinary coordination for proper diagnosis and management. The pathobiology of MPN-SVT is not fully understood, but recent developments have revealed the central role of endothelial cells. In this multidisciplinary review, we summarize the epidemiology of MPN-SVT and then critically evaluate the pathogenic features of this complication, with a focus on endothelial cell biology. We then discuss diagnostic considerations, including imaging modalities and MPN-specific investigations. Finally, we critically review the evidence supporting clinical management of MPN-SVT, including anticoagulation, interventional radiology procedures, MPN-related therapies, and liver transplantation. We conclude that further studies are needed to improve our understanding of MPN-SVT and the outcomes of patients with this debilitating complication., Competing Interests: Declaration of competing interest DT receives research clinical research funding from Astellas Pharma and consulting fees from AbbVie and CTI Biopharma. JDB receives research funding from Bayer independent of studies herein. RP receives consultancy fees from Sirtex Medical and is on the advisory boards of Medtronic, Blackswan Medical, and Scientia Inc. and is on the speakers' bureau of Medtronic. JM receives clinical research funding from Incyte, Janssen, CTI Biopharma, PharmaEssentia, Novartis, Roche, Kartos, Promedior Merck and consulting fees from Promedior, Prelude, Galecto, Incyte, Celgene, Kartos, and Geron. The rest of the authors have no financial conflicts of interest to disclose., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
33. The Quantification and Significance of Extramedullary Hematopoiesis Seen on Liver Biopsy Specimens.
- Author
-
Tremblay D, Saberi S, Mascarenhas J, Schiano TD, and Fiel MI
- Subjects
- Adult, Biopsy, Humans, Liver, Hematologic Diseases, Hematopoiesis, Extramedullary, Myeloproliferative Disorders
- Abstract
Objectives: Extramedullary hematopoiesis (EMH) may occur in the liver and is often considered pathologic in adults. Many hematologic and nonhematologic disorders are associated with the development of EMH. However, it is unclear whether the presence of EMH is always pathologic. At present, no formal grading system for EMH in the liver exists., Methods: We reviewed 42 liver biopsy specimens with EMH and developed a novel grading system to quantify the degree of EMH from 1 to 3 based on the number of EMH foci in 10 high-power fields., Results: Most patients had nonhematologic conditions (n = 25). Seventeen patients had a hematologic condition, most frequently a myeloproliferative neoplasm (n = 9). Patients with an underlying hematologic condition had a significantly higher EMH grade compared with those without a hematologic condition (P < .0001). All patients with grade 3 EMH had an underlying hematologic diagnosis, and most (86%) patients with grade 1 EMH had a nonhematologic disorder., Conclusions: Our data suggest that EMH grading in the liver is feasible and can identify patients who may have an underlying hematologic condition, which can guide further diagnostic workup., (© The Author(s) 2022. Published by Oxford University Press on behalf of American Society for Clinical Pathology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
34. Anagrelide for platelet-directed cytoreduction in polycythemia vera: Insights into utility and safety outcomes from a large multi-center database.
- Author
-
Rippel N, Tremblay D, Zubizarreta N, Podoltsev N, Gotlib J, Heaney M, Kuykendall A, O'Connell C, Shammo JM, Fleischman A, Kremyanskaya M, Hoffman R, Mesa R, Yacoub A, and Mascarenhas J
- Subjects
- Cytoreduction Surgical Procedures, Humans, Quinazolines, Polycythemia Vera drug therapy, Thrombocythemia, Essential drug therapy, Thrombocytosis therapy, Thrombosis
- Abstract
Anagrelide (ANA) is a platelet-specific cytoreductive agent utilized in the guideline-directed management of high-risk essential thrombocythemia. In the context of polycythemia vera (PV), ANA is occasionally employed in clinical practice, although data has not consistently demonstrated a benefit to targeting a platelet goal as a therapeutic endpoint. The aim of the current study was to delineate the patterns of ANA use in PV, and to describe outcomes and toxicities. Within a multi-center cohort of 527 patients with PV, 48 received ANA (9 excluded for absent data). 27 (69.2%) had high-risk PV, 10 (25.6%) had prior thrombosis, and none had extreme thrombocytosis, acquired von Willebrand disease, and/or documented resistance to hydroxyurea. While ANA effectively lowered median platelet count, 43.5% of patients had an unresolved thrombocytosis at time of ANA discontinuation. Treatment-emergent adverse events-including headaches, cardiac palpitations and arrhythmias, nausea, vomiting and/or diarrhea-led to ANA discontinuation in 76.9% of patients. Further, three patients experienced arterial thromboses during a median duration of 27.5 months of ANA therapy. In conclusion, this study highlights ANA's restrictive tolerability profile which, compounded by the absence of clear advantage to strict platelet control in PV, suggests the use of ANA should be limited in this setting., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
35. Thrombocytopenia in Patients With Myelofibrosis: Pathogenesis, Prevalence, Prognostic Impact, and Treatment.
- Author
-
Sastow D, Mascarenhas J, and Tremblay D
- Subjects
- Humans, Nitriles, Prevalence, Prognosis, Protein Kinase Inhibitors adverse effects, Pyrazoles adverse effects, Pyrimidines adverse effects, Splenomegaly etiology, Anemia chemically induced, Primary Myelofibrosis complications, Primary Myelofibrosis diagnosis, Primary Myelofibrosis therapy, Thrombocytopenia epidemiology, Thrombocytopenia etiology, Thrombocytopenia therapy
- Abstract
Myelofibrosis (MF) is a clonal hematopoietic stem cell neoplasm, characterized by pathologic myeloproliferation associated with inflammatory and pro-angiogenic cytokine release, that results in functional compromise of the bone marrow. Thrombocytopenia is a disease-related feature of MF, which portends a poor prognosis impacting overall survival (OS) and leukemia free survival. Thrombocytopenia in MF has multiple causes including ineffective hematopoiesis, splenic sequestration, and treatment-related effects. Presently, allogeneic hematopoietic stem cell transplantation (HSCT) remains the only curable treatment for MF, which, unfortunately, is only a viable option for a minority of patients. All other currently available therapies are either focused on improving cytopenias or the alleviating systemic symptoms and burdensome splenomegaly. While JAK2 inhibitors have moved to the forefront of MF therapy, available JAK inhibitors are advised against in patients with severe thrombocytopenia (platelets < 50 × 10
9 /L). In this review, we describe the pathogenesis, prevalence, and prognostic significance of thrombocytopenia in MF. We also explore the value and limitations of treatments directed at addressing cytopenias, splenomegaly and symptom burden, and those with potential disease modification. We conclude by proposing a treatment algorithm for patients with MF and severe thrombocytopenia., Competing Interests: Declaration of Competing Interest DS has no conlicts of interest; JM receives research funding paid to the institution from Incyte, Novartis, Roche, Merck, Geron, CTI Bio, BMS, Celgene, Kartos, PharmaEssentia and consulting fees from Incyte, CTI Bio, Constellation, Geron, Kartos, BMS, Celgene, Novartis, Karyopharm, Sierra Oncology, PharmaEssentia, and Galecto; DT receives contracted research funding paid to his institution from Astellas Pharma and consulting fees from AbbVie and CTI Biopharma., (Copyright © 2022. Published by Elsevier Inc.)- Published
- 2022
- Full Text
- View/download PDF
36. Male-specific late effects in adult hematopoietic cell transplantation recipients: a systematic review from the Late Effects and Quality of Life Working Committee of the Center for International Blood and Marrow Transplant Research and Transplant Complications Working Party of the European Society of Blood and Marrow Transplantation.
- Author
-
Phelan R, Im A, Hunter RL, Inamoto Y, Lupo-Stanghellini MT, Rovo A, Badawy SM, Burns L, Eissa H, Murthy HS, Prasad P, Sharma A, Suelzer E, Agrawal V, Aljurf M, Baker K, Basak GW, Buchbinder D, DeFilipp Z, Grkovic LD, Dias A, Einsele H, Eisenberg ML, Epperla N, Farhadfar N, Flatau A, Gale RP, Greinix H, Hamilton BK, Hashmi S, Hematti P, Jamani K, Maharaj D, Murray J, Naik S, Nathan S, Pavletic S, Peric Z, Pulanic D, Ross R, Salonia A, Sanchez-Ortega I, Savani BN, Schechter T, Shah AJ, Smith SM, Snowden JA, Steinberg A, Tremblay D, Vij SC, Walker L, Wolff D, Yared JA, Schoemans H, and Tichelli A
- Subjects
- Adult, Bone Marrow, Disease Progression, Humans, Male, Quality of Life, Transplant Recipients, Graft vs Host Disease etiology, Hematopoietic Stem Cell Transplantation adverse effects
- Abstract
Male-specific late effects after hematopoietic cell transplantation (HCT) include genital chronic graft-versus-host disease (GvHD), hypogonadism, sexual dysfunction, infertility, and subsequent malignancies. They may be closely intertwined and cause prolonged morbidity and decreased quality of life after HCT. We provide a systematic review of male-specific late effects in a collaboration between transplant physicians, endocrinologists, urologists, dermatologists, and sexual health professionals through the Late Effects and Quality of Life Working Committee of the Center for International Blood and Marrow Transplant Research, and the Transplant Complications Working Party of the European Society of Blood and Marrow Transplantation. The systematic review summarizes incidence, risk factors, screening, prevention and treatment of these complications and provides consensus evidence-based recommendations for clinical practice and future research., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
37. The Effect of Fedratinib, A Selective Inhibitor of Janus Kinase 2, on Weight and Metabolic Parameters in Patients with Intermediate- or High-risk Myelofibrosis.
- Author
-
Tremblay D, Cavalli L, Sy O, Rose S, and Mascarenhas J
- Subjects
- Humans, Janus Kinase 1, Janus Kinase 2, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Pyrrolidines, Sulfonamides, Primary Myelofibrosis drug therapy
- Published
- 2022
- Full Text
- View/download PDF
38. Momelotinib for the treatment of myelofibrosis with anemia.
- Author
-
Tremblay D and Mesa R
- Subjects
- Benzamides therapeutic use, Humans, Janus Kinase 2 genetics, Nitriles therapeutic use, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Pyrimidines, Quality of Life, Anemia drug therapy, Anemia etiology, Janus Kinase Inhibitors, Primary Myelofibrosis complications, Primary Myelofibrosis drug therapy, Primary Myelofibrosis metabolism
- Abstract
Myelofibrosis is a myeloproliferative neoplasm characterized by splenomegaly, debilitating constitutional symptoms and bone marrow failure. Disease-related anemia is common and associated with an inferior quality of life and survival. Unfortunately, few therapies exist to improve hemoglobin in myelofibrosis patients. Momelotinib is a JAK1/JAK2 inhibitor that also antagonizes ACVR1, leading to downregulation of hepcidin expression and increased availability of iron for erythropoiesis. In clinical testing, momelotinib has demonstrated a unique ability to improve hemoglobin and reduce transfusion burden in myelofibrosis patients with baseline anemia, while producing reductions in spleen size and symptom burden. This review explores the preclinical rationale, clinical trial data and future role of momelotinib in the evolving therapeutic landscape of myelofibrosis.
- Published
- 2022
- Full Text
- View/download PDF
39. Addressing symptom burden in myeloproliferative neoplasms.
- Author
-
Tremblay D and Mesa R
- Subjects
- Humans, Quality of Life, Myeloproliferative Disorders diagnosis, Myeloproliferative Disorders therapy, Neoplasms
- Abstract
Patients with myeloproliferative neoplasms (MPNs) suffer from often debilitating constitutional symptoms that negatively impact quality of life to a degree similar to patients with metastatic solid tumors. Despite heterogeneity in the breadth and severity of symptoms in MPNs, research into symptom assessment has led to the creation of well validated patient reported outcome tools, including the Myeloproliferative Neoplasm Symptom Assessment Form Total Symptom Score. Currently available pharmacologic therapies, particularly JAK inhibitors, result in substantial reduction in symptom burden for patients with myelofibrosis, as well as select patient with polycythemia vera and essential thrombocythemia. Non-pharmacologic therapies including yoga and meditation have also been investigated. In this review, we focus on the pathogenesis and assessment of constitutional symptoms in MPNs. We detail currently available therapies to address symptom burden and highlight several novel agents in development. We end by discussing unmet needs and exploring the future of symptom assessment and treatment in MPNs., Competing Interests: Declaration of competing interest Douglas Tremblay receives contracted research funding paid to his institution from Astellas Pharma and consulting fees from AbbVie. Ruben Mesa receives contracted research funding paid to his institution from Celgene, Incyte, AbbVie, Samus, Genotech, Promedior, CTI Biopharma, Constellation and the Mays Cancer Center P30 Cancer Center Support Grant from the National Cancer Institute (CA054174). He receives consulting fees from Novartis, Sierra Oncology, LaJolla Pharmaceutical, and Constellation., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
40. Contemporary and future strategies in polycythemia vera.
- Author
-
Waksal JA and Tremblay DA
- Subjects
- Humans, Janus Kinase 2 genetics, Hemorrhage, Polycythemia Vera genetics, Polycythemia Vera therapy, Polycythemia Vera diagnosis, Primary Myelofibrosis etiology
- Abstract
Polycythemia vera (PV) is characterized by clonal proliferation of a hematopoietic stem cell leading to erythrocytosis. Patients with PV have significantly higher morbidity and mortality compared to the general population due to increased risk of thrombosis, hemorrhage, and well-characterized microvascular and constitutional symptoms. There is also a propensity to transform to myelofibrosis and to an aggressive form of acute leukemia, further increasing morbidity and mortality. Current management is aimed at reducing the risk of thromboembolic events and improving symptom burden; however, none of the existing therapies have proven the ability to deplete the underlying malignant clone, or definitively reduce the risk of disease, progression leaving a large area of unmet need. In this review, we highlight the pathophysiology of PV, current management and limitations therein. We propose highly debated clinical practices that require further investigation. We conclude by discussing therapies in development and how these may fill unmet needs and be incorporated into the future PV treatment paradigm., Competing Interests: Declaration of competing interest JW reports no conflicts of interest. DT receives contracted research funding paid to his institution from Astellas Pharmaceuticals and consulting fees from AbbVie., (Copyright © 2022. Published by Elsevier Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
41. Male-Specific Late Effects in Adult Hematopoietic Cell Transplantation Recipients: A Systematic Review from the Late Effects and Quality of Life Working Committee of the Center for International Blood and Marrow Transplant Research and Transplant Complications Working Party of the European Society of Blood and Marrow Transplantation.
- Author
-
Phelan R, Im A, Hunter RL, Inamoto Y, Lupo-Stanghellini MT, Rovo A, Badawy SM, Burns L, Eissa H, Murthy HS, Prasad P, Sharma A, Suelzer E, Agrawal V, Aljurf M, Baker K, Basak GW, Buchbinder D, DeFilipp Z, Grkovic LD, Dias A, Einsele H, Eisenberg ML, Epperla N, Farhadfar N, Flatau A, Gale RP, Greinix H, Hamilton BK, Hashmi S, Hematti P, Jamani K, Maharaj D, Murray J, Naik S, Nathan S, Pavletic S, Peric Z, Pulanic D, Ross R, Salonia A, Sanchez-Ortega I, Savani BN, Schechter T, Shah AJ, Smith SM, Snowden JA, Steinberg A, Tremblay D, Vij SC, Walker L, Wolff D, Yared JA, Schoemans H, and Tichelli A
- Subjects
- Adult, Bone Marrow, Female, Humans, Male, Quality of Life, Graft vs Host Disease epidemiology, Hematopoietic Stem Cell Transplantation adverse effects, Hypogonadism epidemiology, Infertility etiology, Testicular Neoplasms etiology
- Abstract
Male-specific late effects after hematopoietic cell transplantation (HCT) include genital chronic graft-versus-host disease (GVHD), hypogonadism, sexual dysfunction, infertility, and subsequent malignancies, such as prostate, penile, and testicular cancer. These effects may be closely intertwined and cause prolonged morbidity and decreased quality of life after HCT. Here we provide a systematic review of male-specific late effects in a collaboration among transplantation physicians, endocrinologists, urologists, dermatologists, and sexual health professionals through the Late Effects and Quality of Life Working Committee of the Center for International Blood and Marrow Transplant Research and the Transplant Complications Working Party of the European Society of Blood and Marrow Transplantation. We used a systematic review methodology to summarize incidence, risk factors, screening, prevention, and treatment of these complications and provide consensus evidence-based recommendations for clinical practice and future research. Most of the evidence regarding male GVHD is still based on limited data, precluding strong therapeutic recommendations. Therefore, we recommend systematic screening for male genital GVHD regularly and reporting of cases to large registries to allow for a better understanding. Future research also should address treatment, given the little published evidence currently available. Male-specific endocrine consequences of HCT include hypogonadism, which also may affect bone health. Given the scanty evidence, current recommendations for hormone substitution and/or bone health treatment are based on similar principles as for the general population. Following HCT, sexual health decreases, and this topic should be addressed at regular intervals. Future studies should focus on interventional strategies to address sexual dysfunction. Infertility remains prevalent in patients having undergone myeloablative conditioning, warranting the offer of sperm preservation for all HCT candidates. Most studies on fertility rely on descriptive registry analysis and surveys, underscoring the importance of reporting post-HCT conception data to large registries. Although the quality of evidence is low, the development of cancer in male genital organs does not seem more prevalent in HCT recipients compared with the general population; however, subsequent malignancies in general seem to be more prevalent in males than in females, and special attention should be given to skin and oral mucosa. Male-specific late effects, which likely are more underreported than female-specific complications, should be systematically considered during the regular follow-up visits of male survivors who have undergone HCT. Care of patients with male-specific late effects warrants close collaboration between transplantation physicians and specialists from other involved disciplines. Future research should be directed toward better data collection on male-specific late effects and on studies about the interrelationships among these late effects, to allow the development of evidence-based effective management practices., (Copyright © 2022 The American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
42. European LeukemiaNet Response Predicts Disease Progression but Not Thrombosis in Polycythemia Vera.
- Author
-
Tremblay D, Srisuwananukorn A, Ronner L, Podoltsev N, Gotlib J, Heaney ML, Kuykendall A, O'Connell CL, Shammo JM, Fleischman A, Mesa R, Yacoub A, Hoffman R, Moshier E, Zubizarreta N, and Mascarenhas J
- Published
- 2022
- Full Text
- View/download PDF
43. Low-risk polycythemia vera and essential thrombocythemia: management considerations and future directions.
- Author
-
Goulart H, Mascarenhas J, and Tremblay D
- Subjects
- Aspirin therapeutic use, Humans, Quality of Life, Myeloproliferative Disorders diagnosis, Polycythemia Vera diagnosis, Polycythemia Vera epidemiology, Polycythemia Vera therapy, Primary Myelofibrosis diagnosis, Thrombocythemia, Essential drug therapy, Thrombocythemia, Essential therapy, Thrombosis complications, Thrombosis prevention & control
- Abstract
Thrombotic events are a distinctive feature of the myeloproliferative neoplasms (MPNs) polycythemia vera (PV) and essential thrombocythemia (ET). Patients with these MPNs may also experience a poor quality of life secondary to symptom burden, as well as progression of disease to acute leukemia or myelofibrosis. Over the years, various risk stratification methods have evolved in order to attempt to predict thrombotic risk, which is the largest contributor of morbidity and mortality in these patients. More than half of PV and ET patients are low- or intermediate-risk disease status at the time of diagnosis. While therapeutic development is presently focused on high-risk patients, there is a paucity of therapies, outside of aspirin and therapeutic phlebotomy, which can reduce the thrombotic risk or delay disease progression in low-risk patients. In this review, we first describe the various complications that patients with PV and ET experience, and then detail our evolving understanding of risk stratification in these diseases. We then highlight the available evidence on the management of low-risk PV and ET and include a description of novel therapies currently under investigation in this space. We conclude with recommendations for future directions to advance our understanding and improve the treatment of low-risk PV and ET., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
44. Novel treatments for myelofibrosis: beyond JAK inhibitors.
- Author
-
Tremblay D and Mesa R
- Subjects
- Humans, Janus Kinases metabolism, Janus Kinases therapeutic use, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, STAT Transcription Factors metabolism, STAT Transcription Factors therapeutic use, Signal Transduction, Splenomegaly complications, Janus Kinase Inhibitors pharmacology, Janus Kinase Inhibitors therapeutic use, Primary Myelofibrosis pathology
- Abstract
Myelofibrosis is a chronic hematologic malignancy characterized by constitutional symptoms, bone marrow fibrosis, extramedullary hematopoiesis resulting in splenomegaly and a propensity toward leukemic progression. Given the central role of the JAK-STAT pathway in the pathobiology of myelofibrosis, JAK inhibitors are the mainstay of current pharmacologic management. Although these therapies have produced meaningful improvements in splenomegaly and symptom burden, JAK inhibitors do not significantly impact disease progression. In addition, many patients are ineligible because of disease-related cytopenias, which are exacerbated by JAK inhibitors. Therefore, there is a continued effort to identify targets outside the JAK-STAT pathway. In this review, we discuss novel therapies in development for myelofibrosis. We focus on the preclinical rationale, efficacy and safety data for non-JAK inhibitor therapies that have published or presented clinical data. Specifically, we discuss agents that target epigenetic modification (pelabresib, bomedemstat), apoptosis (navitoclax, navtemdalin), signaling pathways (parsaclisib), bone marrow fibrosis (AVID200, PRM-151), in addition to other targets including telomerase (imetelstat), selective inhibitor of nuclear transport (selinexor), CD123 (tagraxofusp) and erythroid maturation (luspatercept). We end by providing commentary on the ongoing and future therapeutic development in myelofibrosis., (© 2022. Japanese Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
45. Myelodysplastic syndrome/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis: Ringing in a new future.
- Author
-
Nathan DI, Feld J, El Jamal SM, Mascarenhas J, and Tremblay D
- Subjects
- Humans, Mutation, RNA Splicing Factors genetics, Syndrome, Anemia, Sideroblastic genetics, Anemia, Sideroblastic pathology, Anemia, Sideroblastic therapy, Hematologic Neoplasms complications, Myelodysplastic-Myeloproliferative Diseases complications, Myelodysplastic-Myeloproliferative Diseases genetics, Myelodysplastic-Myeloproliferative Diseases therapy, Myeloproliferative Disorders complications, Thrombocytosis genetics, Thrombocytosis therapy
- Abstract
Myelodysplastic syndrome/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T) is a rare hematologic malignancy belonging to the category of myelodysplastic syndrome (MDS)/myeloproliferative neoplasm (MPN) overlap syndromes. While certain clinical features, including anemia and thrombocytosis, are common to both the MDS and MPN disease components, the biologic consequences of the spliceosome mutation SF3B1 results in notable clinical exceptions. Importantly, both overall and leukemia free survival are shorter for MDS/MPN-RS-T when compared to essential thrombocythemia (ET). In the case of MDS/MPN-RS-T, thrombotic risk is not associated with the presence of JAK2V617F, nor history of prior thrombosis, but is associated with the presence of the mutated spliceosome gene SF3B1. In this review, we highlight the biology, pathology, risk stratification, and treatment approach to MDS/MPN-RS-T. In particular, we focus on clinical management concepts, which are largely borrowed from MDS and MPN, including the use of cytoreduction, bone marrow stimulating agents, and the role of allogeneic stem cell transplantation. We end by highlighting unmet needs and future research priorities in MDS/MPN-RS-T., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
46. Combined Drug Targeting of p53-dependent and -independent Pathways Depletes Myelofibrosis Hematopoietic Stem/Progenitor Cells.
- Author
-
Lu M, Xia L, Elmansy N, Clementelli C, Tremblay D, and Hoffman R
- Subjects
- Cells, Cultured, Drug Delivery Systems, Hematopoietic Stem Cells metabolism, Humans, Primary Myelofibrosis metabolism, Signal Transduction drug effects, Antineoplastic Agents pharmacology, Hematopoietic Stem Cells drug effects, Imidazoles pharmacology, Imidazolines pharmacology, Primary Myelofibrosis drug therapy, Pyridines pharmacology, Pyrimidines pharmacology, Tumor Suppressor Protein p53 metabolism
- Abstract
Current therapy for myelofibrosis (MF) results in a limited prolongation of patient survival. In order to improve treatment outcomes, we developed a strategy to effectively deplete MF hematopoietic stem/progenitor cells (HSPCs). In the present study, an imipridone, ONC201, was combined with RG7112, an antagonist of MDM2, a p53 negative regulator, to activate downstream events of the p53 and TNF-related apoptosis-inducing ligand (TRAIL)/death receptor (DR) pathways. As compared to treatment with the individual drugs, the combination of ONC201 and RG7112 promoted greater degrees of apoptosis of MF CD34
+ cells through activation of both p53-dependent and -independent pathways. Importantly, treatment with ONC201-RG7112 not only decreased the number of JAK2V617F+ and calreticulin mutated colonies assayed from MF CD34+ cells, but allowed for the persistence or appearance of JAK2 wild type colonies. Treatment with ONC201 combined with RG7112 could be a potentially effective strategy for treating MF patients., (© 2021. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
47. Emerging drugs for the treatment of myelofibrosis: phase II & III clinical trials.
- Author
-
Tremblay D and Hoffman R
- Subjects
- Humans, Janus Kinase 2, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Janus Kinase Inhibitors pharmacology, Janus Kinase Inhibitors therapeutic use, Pharmaceutical Preparations, Primary Myelofibrosis drug therapy
- Abstract
Introduction: Myelofibrosis is a clonal hematologic malignancy with clinical manifestations that include cytopenias, debilitating constitutional symptoms, splenomegaly, bone marrow fibrosis and a propensity toward leukemic progression. While allogeneic hematopoietic stem cell transplantation can be curative, this therapy is not available for the majority of patients. Ruxolitinib and fedratinib are approved JAK2 inhibitors that have produced meaningful benefits in terms of spleen reduction and symptom improvement, but there remain several unmet needs., Areas Covered: We discuss novel therapies based upon published data from phase II or III clinical trials. Specifically, we cover novel JAK inhibitors (momelotinib and pacritinib), and agents that target bromodomain and extra-terminal domain (pelabresib), the antiapoptotic proteins BCL-2/BCL-xL (navitoclax), MDM2 (navtemadlin), phosphatidylinositol 3-kinase (parsaclisib), or telomerase (imetelstat)., Expert Opinion: Patients with disease related cytopenias are ineligible for currently approved JAK2 inhibitors. However, momelotinib and pacritinib may be able to fill this void. Novel therapies are being evaluated in the upfront setting to improve the depth and duration of responses with ruxolitinib. Future evaluation of agents must be judged on their potential to modify disease progression, which current JAK2 inhibitors lack. Combination therapy, possibly with an immunotherapeutic agent might serve as key components of future myelofibrosis treatment options.
- Published
- 2021
- Full Text
- View/download PDF
48. Direct oral anticoagulants for myeloproliferative neoplasms: results from an international study on 442 patients.
- Author
-
Barbui T, De Stefano V, Carobbio A, Iurlo A, Alvarez-Larran A, Cuevas B, Ferrer Marín F, Vannucchi AM, Palandri F, Harrison C, Sibai H, Griesshammer M, Bonifacio M, Elli EM, Trotti C, Koschmieder S, Carli G, Benevolo G, Ianotto JC, Goel S, Falanga A, Betti S, Cattaneo D, Arellano-Rodrigo E, Mannelli L, Vianelli N, Doyle A, Gupta V, Wille K, Tremblay D, and Mascarenhas J
- Subjects
- Administration, Oral, Atrial Fibrillation etiology, Atrial Fibrillation pathology, Humans, International Agencies, Venous Thromboembolism etiology, Venous Thromboembolism pathology, Anticoagulants administration & dosage, Atrial Fibrillation drug therapy, Myeloproliferative Disorders complications, Venous Thromboembolism drug therapy
- Published
- 2021
- Full Text
- View/download PDF
49. Care disruptions among patients with lung cancer: A COVID-19 and cancer outcomes study.
- Author
-
Bhalla S, Bakouny Z, Schmidt AL, Labaki C, Steinharter JA, Tremblay DA, Awad MM, Kessler AJ, Haddad RI, Evans M, Busser F, Wotman M, Curran CR, Zimmerman BS, Bouchard G, Jun T, Nuzzo PV, Qin Q, Hirsch L, Feld J, Kelleher KM, Seidman D, Huang HH, Anderson-Keightly HM, El Zarif T, Abou Alaiwi S, Rosenbloom TD, Stewart PS, Galsky MD, Choueiri TK, and Doroshow DB
- Subjects
- COVID-19 Testing, Humans, Pandemics, Prospective Studies, SARS-CoV-2, COVID-19, Lung Neoplasms epidemiology, Lung Neoplasms therapy
- Abstract
Introduction: Patients with lung cancer (LC) are susceptible to severe outcomes from COVID-19. This study evaluated disruption to care of patients with LC during the COVID-19 pandemic., Methods: The COVID-19 and Cancer Outcomes Study (CCOS) is a prospective cohort study comprised of patients with a current or past history of hematological or solid malignancies with outpatient visits between March 2 and March 6, 2020, at two academic cancer centers in the Northeastern United States (US). Data was collected for the three months prior to the index week (baseline period) and the following three months (pandemic period)., Results: 313 of 2365 patients had LC, 1578 had other solid tumors, and 474 had hematological malignancies. Patients with LC were not at increased risk of COVID-19 diagnosis compared to patients with other solid or hematological malignancies. When comparing data from the pandemic period to the baseline period, patients with LC were more likely to have a decrease in in-person visits compared to patients with other solid tumors (aOR 1.94; 95% CI, 1.46-2.58), but without an increase in telehealth visits (aOR 1.13; 95% CI 0.85-1.50). Patients with LC were more likely to experience pandemic-related treatment delays than patients with other solid tumors (aOR 1.80; 95% CI 1.13-2.80) and were more likely to experience imaging/diagnostic procedure delays than patients with other solid tumors (aOR 2.59; 95% CI, 1.46-4.47) and hematological malignancies (aOR 2.01; 95% CI, 1.02-3.93). Among patients on systemic therapy, patients with LC were also at increased risk for decreased in-person visits and increased treatment delays compared to those with other solid tumors., Discussion: Patients with LC experienced increased cancer care disruption compared to patients with other malignancies during the early phase of the COVID-19 pandemic. Focused efforts to ensure continuity of care for this patient population are warranted., (Copyright © 2021. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
50. The utility and complications of plasma administration in cirrhotic patients undergoing minimally invasive procedures.
- Author
-
Diaz KE, Tremblay D, Ozturk B, Ezaz G, Arinsburg S, Jhang J, and Schiano TD
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Retrospective Studies, Blood Component Transfusion adverse effects, Hemorrhage prevention & control, Liver Cirrhosis therapy, Minimally Invasive Surgical Procedures adverse effects, Plasma chemistry
- Abstract
Patients with cirrhosis have coagulopathy often necessitating correction with blood products, such as plasma products (fresh frozen plasma and plasma frozen within 24 h) prior to certain invasive procedures. However, plasma administration has the potential for substantial negative adverse effects such as volume overload, transfusion-related lung injury and allergic/anaphylactic reactions. In addition, its effectiveness in preventing bleeding is similarly unclear. The purpose of this study was to determine the safety and efficacy of plasma administration in cirrhotic patients undergoing minimally invasive procedures, specifically vascular access placement, transjugular liver biopsies, renal biopsies and thoracenteses. In this retrospective cohort study, we identified patients receiving plasma products in preparation for an invasive procedure, with the primary outcomes of volume overload and bleeding. Of the 145 transfusion events that met the criteria from 2015 to 2018, the median INR decreased from 2.7 to 2.2 pre and post plasma administration and 13.8% of recipients had complications of volume overload. The cost of acquisition of plasma administered below clinically impactful doses accumulates to an estimated 19 000 dollars over this time period, not including nursing preparation or production costs. Plasma products minimally, if at all, improved laboratory values of coagulation and in some patients led to adverse effects., (Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.