54 results on '"Geyer MB"'
Search Results
2. Real-world incidence and prevention of tumor lysis syndrome in chronic lymphocytic leukemia treated with venetoclax.
- Author
-
Valtis YK, Nemirovsky D, Derkach A, Sharan S, Kabel C, Ortiz R, Thompson MC, Roeker LE, and Geyer MB
- Abstract
Abstract: Venetoclax is a B-cell lymphoma 2 inhibitor used in chronic lymphocytic leukemia (CLL), which can cause tumor lysis syndrome (TLS). We aimed to determine the incidence of, and risk factors for, TLS among patients with CLL/small lymphocytic lymphoma who received treatment with venetoclax at our institution from 1 January 2016 to 31 December 2020. We included 616 venetoclax escalations among 136 patients with CLL. Overall, 74 patients (54%) underwent escalation exclusively outpatient, 35 (26%) had at least 1 planned hospitalization, and 27 (20%) were escalated exclusively inpatient. During venetoclax initiation, 86% of patients received allopurinol, 71% intravenous hydration, 18% phosphate binders, and 10% prophylactic rasburicase. Among the entire cohort, 7 patients (5.1%) developed laboratory TLS by modified Cairo Bishop criteria and none developed clinical TLS. Incidence of laboratory TLS was 15% for those escalated exclusively inpatient, 2.9% for those with any prophylactic hospitalization, and 2.7% for those escalated exclusively outpatient. Those who developed TLS were more likely to have higher TLS risk, and no additional risk factors were identified. In this single institution retrospective cohort study, laboratory TLS was observed, although clinical TLS was not. Prophylactic measures, including use of IV hydration, may have contributed to low rates of observed TLS in the outpatient setting., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. HMA/VEN treatment modifications and associated outcomes in IDH -mutant AML.
- Author
-
Chin KK, Derkach A, Famulare C, Gupta GK, Borge PD, Geyer MB, Goldberg AD, Haque T, Park JH, Roeker LE, Tallman MS, Stahl M, and Stein EM
- Abstract
Hypomethylating agents (HMA) and venetoclax (VEN) are commonly used in patients with IDH -mutated ( IDH m) acute myeloid leukemia (AML) ineligible for induction chemotherapy. While prior studies demonstrated high response and survival rates with HMA/VEN in IDH m AML, the impact of treatment modifications in real-world settings is unclear. We retrospectively reviewed 89 IDH m AML patients treated with HMA/VEN from January 2018 to June 2023. CR/CRi rates were 76% in newly diagnosed (ND) and 55% in relapsed/refractory (R/R) patients, and median overall survival was 29.2 months (ND) and 17.1 months (R/R), respectively. Treatment modifications were common. Early VEN reductions were associated with lower response rates but not worse survival. Prolonged cycles were not associated with worse response rates or survival. Significant neutropenia and ED visits or unplanned hospitalizations were considerable before and after CR/CRi, though febrile neutropenia decreased afterward. HMA/VEN is efficacious, with treatment modifications not affecting survival, though long-term toxicities are notable.
- Published
- 2024
- Full Text
- View/download PDF
4. Universal germline genetic testing in patients with hematologic malignancies using DNA isolated from nail clippings.
- Author
-
Ceyhan-Birsoy O, Fiala E, Rana S, Sheehan M, Kennedy J, Yelskaya Z, Rai V, Li Y, Yang C, Wong D, Rijo I, Casanova J, Somar J, Mehta N, Park H, Ostafi S, Arora K, Padunan A, Ewalt MD, Aypar U, Terraf P, Misyura M, Haque S, Behr GG, Haque T, Sulis M, Geyer MB, Forlenza C, Thompson MC, Carlo M, Latham A, Liu Y, Zehir A, Brannon R, Berger M, Diaz LA Jr, Dogan A, Ladanyi M, Petrova-Drus K, Nafa K, Offit K, Arcila M, Stadler ZK, Walsh MF, and Mandelker D
- Subjects
- Humans, Nails pathology, Male, Female, Hematologic Neoplasms genetics, Hematologic Neoplasms diagnosis, Germ-Line Mutation, Genetic Testing methods
- Published
- 2024
- Full Text
- View/download PDF
5. Atrial arrhythmias following CAR-chimeric antigen receptor T-cell therapy: Incidence, risk factors and biomarker profile.
- Author
-
Shouval R, Goldman A, Flynn JR, El-Moghraby A, Rehman M, Devlin SM, Corona M, Landego I, Lin RJ, Scordo M, Raj SS, Giralt SA, Palomba ML, Dahi PB, Walji M, Salles G, Nath K, Geyer MB, Park JH, Fein JA, Kosmidou I, Shah GL, Liu JE, Perales MA, and Mahmood SS
- Subjects
- Humans, Male, Female, Middle Aged, Risk Factors, Incidence, Aged, Retrospective Studies, Adult, Biomarkers blood, Lymphoma, Non-Hodgkin therapy, Lymphoma, Non-Hodgkin epidemiology, Lymphoma, Non-Hodgkin immunology, Receptors, Chimeric Antigen, Antigens, CD19 immunology, Immunotherapy, Adoptive adverse effects, Arrhythmias, Cardiac etiology, Arrhythmias, Cardiac therapy
- Abstract
Recent reports have raised concerns about the association of chimeric antigen receptor T cell (CAR-T) with non-negligible cardiotoxicity, particularly atrial arrhythmias. First, we conducted a pharmacovigilance study to assess the reporting of atrial arrhythmias following CD19-directed CAR-T. Subsequently, to determine the incidence, risk factors and outcomes of atrial arrhythmias post-CAR-T, we compiled a retrospective single-centre cohort of non-Hodgkin lymphoma patients. Only commercial CAR-T products were considered. Atrial arrhythmias were nearly fourfold more likely to be reported after CAR-T therapy compared to all other cancer patients in the FAERS (adjusted ROR = 3.76 [95% CI 2.67-5.29]). Of the 236 patients in our institutional cohort, 23 (10%) developed atrial arrhythmias post-CAR-T, including 12 de novo arrhythmias, with most (83%) requiring medical intervention. Atrial arrhythmias frequently co-occurred with cytokine release syndrome and were associated with higher post-CAR-T infusion peak levels of IL-10, TNF-alpha and LDH, and lower trough levels of fibrinogen. In a multivariable analysis, risk factors for atrial arrhythmia were history of atrial arrhythmia (OR = 6.80 [2.39-19.6]) and using CAR-T product with a CD28-costimulatory domain (OR = 5.17 [1.72-18.6]). Atrial arrhythmias following CD19-CAR-T therapy are prevalent and associated with elevated inflammatory biomarkers, a history of atrial arrhythmia and the use of a CAR-T product with a CD28 costimulatory domain., (© 2024 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
6. Multilineage involvement in KMT2A-rearranged B acute lymphoblastic leukaemia: cell-of-origin, biology, and clinical implications.
- Author
-
Aypar U, Dilip D, Gadde R, Londono DM, Liu Y, Gao Q, Geyer MB, Derkach A, Zhang Y, Glass JL, Roshal M, and Xiao W
- Subjects
- Humans, Female, Child, Male, Child, Preschool, Adolescent, Fusion Proteins, bcr-abl genetics, Adult, In Situ Hybridization, Fluorescence, Infant, Gene Rearrangement, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Young Adult, Middle Aged, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Aged, Myeloid-Lymphoid Leukemia Protein genetics, Histone-Lysine N-Methyltransferase genetics
- Abstract
Aims: B lymphoblastic leukaemia/lymphoma (B-ALL) is thought to originate from Pro/Pre-B cells and the genetic aberrations largely reside in lymphoid-committed cells. A recent study demonstrated that a proportion of paediatric B-ALL patients have BCR::ABL1 fusion in myeloid cells, suggesting a chronic myeloid leukaemia (CML)-like biology in this peculiar subset of B-ALL, although it is not entirely clear if the CD19-negative precursor compartment is a source of the myeloid cells. Moreover, the observation has not yet been extended to other fusion-driven B-ALLs., Methods and Results: In this study we investigated a cohort of KMT2A-rearranged B-ALL patients with a comparison to BCR::ABL1-rearranged B-ALL by performing cell sorting via flow cytometry followed by FISH (fluorescence in situ hybridization) analysis on each of the sorted populations. In addition, RNA sequencing was performed on one of the sorted populations. These analyses showed that (1) multilineage involvement was present in 53% of BCR::ABL1 and 36% of KMT2A-rearranged B-ALL regardless of age, (2) multilineage involvement created pitfalls for residual disease monitoring, and (3) HSPC transcriptome signatures were upregulated in KMT2A-rearranged B-ALL with multilineage involvement., Conclusions: In summary, multilineage involvement is common in both BCR::ABL1-rearranged and KMT2A-rearranged B-ALL, which should be taken into consideration when interpreting the disease burden during the clinical course., (© 2024 John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
7. Acute Febrile Neutrophilic Dermatosis (Sweet Syndrome) in Acute Myeloid Leukemia Patients: A 28-Year Institutional Experience.
- Author
-
Cowen EA, Barrios DM, Pulitzer MP, Moy AP, Dusza SW, De Wolf S, Geyer MB, and Markova A
- Subjects
- Humans, Female, Male, Middle Aged, Retrospective Studies, Aged, Adult, Aged, 80 and over, Incidence, Fever etiology, Sweet Syndrome diagnosis, Sweet Syndrome etiology, Sweet Syndrome pathology, Leukemia, Myeloid, Acute complications, Leukemia, Myeloid, Acute diagnosis
- Abstract
Introduction: Sweet syndrome (SS) is well known to be associated with underlying hematologic malignancies. The incidence and qualities of SS among novel targeted therapies for acute myeloid leukemia (AML) have not yet been described., Methods: Through retrospective review of 19,432 patients diagnosed with acute/chronic leukemia or myelodysplastic syndromes/myeloproliferative neoplasms (MDS+/-MPN) over 28 years, we calculated the incidence of SS in the setting of select hematologic malignancies and described the clinicopathologic characteristics of SS in patients with onset of SS after initiation of novel AML-targeted therapies., Results: Overall incidence of SS was 0.36% (95% CI: 0.27-0.45%), which was significantly higher among patients with AML (50/5,248, 0.94%; 95% CI: 0.71-1.25%). Nine AML patients were on 4 classes of novel targeted treatments - IDH1/2 inhibitor alone, FLT3 inhibitor, IDH2 and DOT1L inhibitor, and anti-CD33 therapy. In therapies inducing myeloid blast differentiation, SS occurred at later onset following treatment., Conclusions: In AML patients with fever and unusual skin lesions, physicians may consider SS earlier, which may shorten time to diagnosis. Future assessments of SS among patients treated with novel therapies for AML and molecular studies of biopsies may help further explain this dermatologic adverse event with earlier diagnosis and management of neutrophilic dermatoses in these patients., (© 2023 S. Karger AG, Basel.)
- Published
- 2024
- Full Text
- View/download PDF
8. Acute myeloid leukemia with mixed phenotype is characterized by stemness transcriptomic signatures and limited lineage plasticity.
- Author
-
Galera P, Dilip D, Derkach A, Chan A, Zhang Y, Persuad S, Mishera T, Liu Y, Famulare C, Gao Q, Mata DA, Arcila M, Geyer MB, Stein E, Dogan A, Levine RL, Roshal M, Glass J, and Xiao W
- Abstract
Mixed phenotype (MP) in acute leukemias poses unique classification and management dilemmas and can be seen in entities other than de novo mixed phenotype acute leukemia (MPAL). Although WHO classification empirically recommends excluding AML with myelodysplasia related changes (AML-MRC) and therapy related AML (t-AML) with mixed phenotype (AML-MP) from MPAL, there is lack of studies investigating the clinical, genetic, and biologic features of AML-MP. We report the first cohort of AML-MRC and t-AML with MP integrating their clinical, immunophenotypic, genomic and transcriptomic features with comparison to MPAL and AML-MRC/t-AML without MP. Both AML cohorts with and without MP shared similar clinical features including adverse outcomes but were different from MPAL. The genomic landscape of AML-MP overlaps with AML without MP but differs from MPAL. AML-MP harbors more frequent RUNX1 mutations than AML without MP and MPAL. RUNX1 mutations did not impact the survival of patients with MPAL. Unsupervised hierarchal clustering based on immunophenotype identified biologically distinct clusters with phenotype/genotype correlation and outcome differences. Furthermore, transcriptomic analysis showed an enrichment for stemness signature in AML-MP and AML without MP as compared to MPAL. Lastly, MPAL but not AML-MP often switched to lymphoid only immunophenotype after treatment. Expression of transcription factors critical for lymphoid differentiation were upregulated only in MPAL, but not in AML-MP. Our study for the first time demonstrates that AML-MP clinically and biologically resembles its AML counterpart without MP and differs from MPAL, supporting the recommendation to exclude these patients from the diagnosis of MPAL. Future studies are needed to elucidate the molecular mechanism of mixed phenotype in AML., Key Points: AML-MP clinically and biologically resembles AML but differs from MPAL. AML-MP shows RUNX1 mutations, stemness signatures and limited lymphoid lineage plasticity.
- Published
- 2023
- Full Text
- View/download PDF
9. Expert consensus guidelines for the prophylaxis and management of tumor lysis syndrome in the United States: Results of a modified Delphi panel.
- Author
-
Perissinotti AJ, Bishop MR, Bubalo J, Geyer MB, Goodrich A, Howard SC, Kula J, Mandayam S, Cairo MS, and Pui CH
- Abstract
Introduction: Tumor lysis syndrome (TLS), which occurs spontaneously or in response to anticancer treatment, results in the release of intracellular potassium, phosphorus, and nucleic acids into the bloodstream, which results in secondary clinical complications that may be fatal. Prior TLS guidelines do not take into consideration potent novel oncologic agents or contemporary treatment paradigms with increased risk of TLS. Thus, a modified Delphi panel of experts was convened to provide an update for TLS management guidelines based upon a combination of supporting literature and practice consensus., Methods: A three-round modified Delphi process was implemented. For round 1, nine expert panelists completed a web-based questionnaire developed using published literature. In round 2, panelists were asked to reconsider their answers to questions that did not reach consensus (defined as ≥ 66% agreement among voting panelists). Round 3 was an unblinded, moderated virtual meeting to discuss any remaining questions that did not reach consensus., Results: Detailed recommendations are given for prophylaxis, monitoring, and management of TLS risks and complications, with hydration being a key element of TLS prophylaxis and management. Guidelines for the management of acute effects of TLS and prevention of long-term renal effects include management of hyperkalemia, hypocalcemia, hyperphosphatemia, and hyperuricemia., Discussion: Although the control of uric acid levels is quite effective with currently available agents, panelists emphasize the importance of monitoring and treating other dangerous electrolyte abnormalities such as hyperkalemia and hyperphosphatemia. Guidelines from this modified Delphi panel should aid clinicians in preventing and managing TLS., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: AJP reports consulting fees from AbbVie, BeiGene, Pfizer, Sanofi-Aventis, and Servier. MRB serves as a member on an advisory board or as a consultant for Autolus Therapeutics, BMS/JUNO Therapeutics, Chimeric Therapeutics, CRISPR Therapeutics, IN8bio, Incyte, Iovance Biotherapeutics, KITE/Gilead, Novartis, PhysIQ, and Sana Biotechnology and is on the speakers’ bureau for ADC Therapeutics, Agios, BMS, Kite/Gilead, Incyte, Sanofi, and Servier. JB reports consulting fees from Sanofi-Aventis. MBG reports research support from Actinium Pharmaceuticals, Amgen, and Sanofi and has served on an advisory board for Sanofi and as a consultant for Novartis. AG reports consulting fees from Sanofi-Aventis. SCH has served as a speaker for Jazz Pharma and Sanofi, has received grant funding from Jazz Pharma, and has served as a consultant for Servier. JK reports consulting fees from Sanofi-Aventis. SM reports grant funding from Horizon; service on advisory boards for Calliditas, Chinook, Travere, and US Renal Care; and service on speaker’s bureaus for Bayer and Calliditas. MSC serves on the speaker’s bureau and as a consultant for Sanofi. C-HP reports support by grant CA21765 from the National Cancer Institute and American Lebanese Syrian Associated Charities, and honorarium from Amgen and Novartis., (Copyright © 2023. Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
10. Interaction between myelodysplasia-related gene mutations and ontogeny in acute myeloid leukemia.
- Author
-
McCarter JGW, Nemirovsky D, Famulare CA, Farnoud N, Mohanty AS, Stone-Molloy ZS, Chervin J, Ball BJ, Epstein-Peterson ZD, Arcila ME, Stonestrom AJ, Dunbar A, Cai SF, Glass JL, Geyer MB, Rampal RK, Berman E, Abdel-Wahab OI, Stein EM, Tallman MS, Levine RL, Goldberg AD, Papaemmanuil E, Zhang Y, Roshal M, Derkach A, and Xiao W
- Subjects
- Humans, Mutation, Prognosis, Risk Factors, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute drug therapy, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes genetics
- Abstract
Accurate classification and risk stratification are critical for clinical decision making in patients with acute myeloid leukemia (AML). In the newly proposed World Health Organization and International Consensus classifications of hematolymphoid neoplasms, the presence of myelodysplasia-related (MR) gene mutations is included as 1 of the diagnostic criteria for AML, AML-MR, based largely on the assumption that these mutations are specific for AML with an antecedent myelodysplastic syndrome. ICC also prioritizes MR gene mutations over ontogeny (as defined in the clinical history). Furthermore, European LeukemiaNet (ELN) 2022 stratifies these MR gene mutations into the adverse-risk group. By thoroughly annotating a cohort of 344 newly diagnosed patients with AML treated at the Memorial Sloan Kettering Cancer Center, we show that ontogeny assignments based on the database registry lack accuracy. MR gene mutations are frequently observed in de novo AML. Among the MR gene mutations, only EZH2 and SF3B1 were associated with an inferior outcome in the univariate analysis. In a multivariate analysis, AML ontogeny had independent prognostic values even after adjusting for age, treatment, allo-transplant and genomic classes or ELN risks. Ontogeny also helped stratify the outcome of AML with MR gene mutations. Finally, de novo AML with MR gene mutations did not show an adverse outcome. In summary, our study emphasizes the importance of accurate ontogeny designation in clinical studies, demonstrates the independent prognostic value of AML ontogeny, and questions the current classification and risk stratification of AML with MR gene mutations., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Dermatologic adverse events in acute lymphocytic leukemia patients treated with bispecific T-cell engager blinatumomab.
- Author
-
Parisi R, Cowen EA, Gu S, Dusza S, Pulitzer M, Geyer MB, King AC, and Markova A
- Subjects
- Humans, T-Lymphocytes, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Lymphoma, Non-Hodgkin drug therapy, Antibodies, Bispecific adverse effects, Antineoplastic Agents adverse effects, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Published
- 2023
- Full Text
- View/download PDF
12. Lenalidomide-associated B-cell ALL: clinical and pathologic correlates and sensitivity to lenalidomide withdrawal.
- Author
-
Geyer MB, Shaffer BC, Bhatnagar B, Mims AS, Klein V, Dilip D, Glass JL, Lozanski G, Hassoun H, Landau H, Zhang Y, Xiao W, Roshal M, and Park JH
- Subjects
- Humans, Aged, Lenalidomide, Retrospective Studies, Progression-Free Survival, Multiple Myeloma therapy, Hematopoietic Stem Cell Transplantation adverse effects, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Burkitt Lymphoma drug therapy
- Abstract
Lenalidomide is an effective component of induction and maintenance therapy for multiple myeloma, though with a risk of secondary malignancies, including acute lymphoblastic leukemia (ALL). In contrast to therapy-related myeloid neoplasia, lenalidomide-associated lymphoblastic neoplasia remains poorly characterized. We conducted a dual institution retrospective study of 32 ALL cases that arose after lenalidomide maintenance (all B-lineage, 31/32 BCR::ABL-negative). B-cell ALL (B-ALL) was diagnosed at median 54 months (range, 5-119) after first exposure to lenalidomide and after median 42 months of cumulative lenalidomide exposure (range, 2-114). High incidence of TP53 mutations (9/19 evaluable cases) and low hypodiploidy (8/26 patients) were identified. Despite median age of 65 years and poor-risk B-ALL features observed in the cohort, rates of complete response (CR) or CR with incomplete hematologic recovery were high (25/28 patients receiving treatment). Median event-free survival was 35.4 months among treated patients (not reached among those undergoing allogeneic hematopoietic cell transplantation [HCT]). Sixteen patients remain alive without evidence of B-ALL after HCT or extended maintenance therapy. We also describe regression of B-ALL or immature B-cell populations with B-ALL immunophenotype after lenalidomide discontinuation in 5 patients, suggesting lenalidomide may drive leukemic progression even after initiation of lymphoblastic neoplasia and that lenalidomide withdrawal alone may be an appropriate first-line intervention in selected patients. Monitoring for early B-ALL-like proliferations may offer opportunities for lenalidomide withdrawal to prevent progression. Established combination chemotherapy regimens, newer surface antigen-targeted approaches, and allogeneic HCT are effective in many patients with lenalidomide-associated B-ALL and should be offered to medically fit patients., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. Biomarkers and cardiovascular outcomes in chimeric antigen receptor T-cell therapy recipients.
- Author
-
Mahmood SS, Riedell PA, Feldman S, George G, Sansoterra SA, Althaus T, Rehman M, Mead E, Liu JE, Devereux RB, Weinsaft JW, Kim J, Balkan L, Barbar T, Lee Chuy K, Harchandani B, Perales MA, Geyer MB, Park JH, Palomba ML, Shouval R, Tomas AA, Shah GL, Yang EH, Gaut DL, Rothberg MV, Horn EM, Leonard JP, Van Besien K, Frigault MJ, Chen Z, Mehrotra B, Neilan TG, and Steingart RM
- Subjects
- Humans, Interleukin-6, Biomarkers, C-Reactive Protein, Troponin, Cell- and Tissue-Based Therapy, Receptors, Chimeric Antigen therapeutic use, Neoplasms, Heart Failure
- Abstract
Aims: Chimeric antigen receptor T-cell therapy (CAR-T) harnesses a patient's immune system to target cancer. There are sparse existing data characterizing death outcomes after CAR-T-related cardiotoxicity. This study examines the association between CAR-T-related severe cardiovascular events (SCE) and mortality., Methods and Results: From a multi-centre registry of 202 patients receiving anti-CD19 CAR-T, covariates including standard baseline cardiovascular and cancer parameters and biomarkers were collected. Severe cardiovascular events were defined as a composite of heart failure, cardiogenic shock, or myocardial infarction. Thirty-three patients experienced SCE, and 108 patients died during a median follow-up of 297 (interquartile range 104-647) days. Those that did and did not die after CAR-T were similar in age, sex, and prior anthracycline use. Those who died had higher peak interleukin (IL)-6 and ferritin levels after CAR-T infusion, and those who experienced SCE had higher peak IL-6, C-reactive protein (CRP), ferritin, and troponin levels. The day-100 and 1-year Kaplan-Meier overall mortality estimates were 18% and 43%, respectively, while the non-relapse mortality (NRM) cumulative incidence rates were 3.5% and 6.7%, respectively. In a Cox model, SCE occurrence following CAR-T was independently associated with increased overall mortality risk [hazard ratio (HR) 2.8, 95% confidence interval (CI) 1.6-4.7] after adjusting for age, cancer type and burden, anthracycline use, cytokine release syndrome grade ≥ 2, pre-existing heart failure, hypertension, and African American ancestry; SCEs were independently associated with increased NRM (HR 3.5, 95% CI 1.4-8.8) after adjusting for cancer burden., Conclusion: Chimeric antigen receptor T-cell therapy recipients who experience SCE have higher overall mortality and NRM and higher peak levels of IL-6, CRP, ferritin, and troponin., Competing Interests: Conflict of interest S.S.M. has received consulting fees from Nektar Therapeutics, Health & Wellness Partners, Medicure. P.A.R. has received consulting fees from AbbVie, BMS, Janssen, Novartis, BeiGene, Kite/Gilead, Intellia Therapeutics, Sana Biotechnology, CVS Caremark, Genmab, Pharmacyclics, Takeda, Karyopharm, Nektar Therapeutics, Nurix Therapeutics, and ADC Therapeutics. M.A.P. reports consulting fees from Adicet, Allovir, Caribou Biosciences, Celgene, Bristol-Myers Squibb, Equilium, Exevir, Incyte, Karyopharm, Kite/Gilead, Merck, Miltenyi Biotec, MorphoSys, Nektar Therapeutics, Novartis, Omeros, OrcaBio, Syncopation, VectivBio AG, and Vor Biopharma, is participating in DSMB of Cidara Therapeutics, Medigene, and Sellas Life Sciences, and is on the scientific advisory board of NexImmune and Omeros. M.B.G. has received institutional grant funding from Sanofi, Amgen, and Actinium and consulting fees from Sanofi, Novartis, and Allogene. M.L.P. has received royalties from Juno and Sers and consulting fees from Novartis, Cellectar, Synthekine, Kite, Seres, Magenta, WindMIL, Rheos, Nektar, Notch, Priothera, Ceramedix, Lygenesis, and Pluto. R.S. has received consulting fees from Mudexus and MyBiotics. G.S. has received research funding from Janssen, Amgen, Beyond Spring, and BMS and is on DSMB of ArcellX. E.H.Y. has received institutional grand funding from CSL Behring, Boehringer Ingelheim, BMS and Eli and Lilly and consulting fees from Pfizer. J.P.L. has received institutional grants from Genentech, Janssen, and Epizyme and consulting fees from Abbvie, Astellas, AstraZeneca, Bayer, Beigene, BMS, Calithera, Constellation, Caribou Biosciences, Eisai, Lilly, Epizyme, Genmab, Grail, Incyte, Jansssen, MEI Pharma, Merck, Mustang Bio, Novartis, Pfizer, Roche/Genentech, Seagen, Second Genome, Sutro, ADC Therapeutics, Miltenyi, and Karyopharm. T.G.N. has received consulting fees from BMS, Genentech, Abbvie, Roche, CRO Oncology, and Sanofi and participates in DSMB of Genentech and received research grant funding from AstraZeneca and BMS., (© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
14. Asciminib monotherapy in patients with CML-CP without BCR::ABL1 T315I mutations treated with at least two prior TKIs: 4-year phase 1 safety and efficacy results.
- Author
-
Mauro MJ, Hughes TP, Kim DW, Rea D, Cortes JE, Hochhaus A, Sasaki K, Breccia M, Talpaz M, Ottmann O, Minami H, Goh YT, DeAngelo DJ, Heinrich MC, Gómez-García de Soria V, le Coutre P, Mahon FX, Janssen JJWM, Deininger M, Shanmuganathan N, Geyer MB, Cacciatore S, Polydoros F, Agrawal N, Hoch M, and Lang F
- Subjects
- Humans, Fusion Proteins, bcr-abl genetics, Mutation, Protein Kinase Inhibitors adverse effects, Drug Resistance, Neoplasm, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Neutropenia chemically induced, Antineoplastic Agents therapeutic use
- Abstract
Asciminib is approved for patients with Philadelphia chromosome-positive chronic-phase chronic myeloid leukemia (CML-CP) who received ≥2 prior tyrosine kinase inhibitors or have the T315I mutation. We report updated results of a phase 1, open-label, nonrandomized trial (NCT02081378) assessing the safety, tolerability, and antileukemic activity of asciminib monotherapy 10-200 mg once or twice daily in 115 patients with CML-CP without T315I (data cutoff: January 6, 2021). After ≈4-year median exposure, 69.6% of patients remained on asciminib. The most common grade ≥3 adverse events (AEs) included increased pancreatic enzymes (22.6%), thrombocytopenia (13.9%), hypertension (13.0%), and neutropenia (12.2%); all-grade AEs (mostly grade 1/2) included musculoskeletal pain (59.1%), upper respiratory tract infection (41.7%), and fatigue (40.9%). Clinical pancreatitis and arterial occlusive events (AOEs) occurred in 7.0% and 8.7%, respectively. Most AEs occurred during year 1; the subsequent likelihood of new events, including AOEs, was low. By data cutoff, among patients without the indicated response at baseline, 61.3% achieved BCR::ABL1 ≤ 1%, 61.6% achieved ≤0.1% (major molecular response [MMR]), and 33.7% achieved ≤0.01% on the International Scale. MMR was maintained in 48/53 patients who achieved it and 19/20 who were in MMR at screening, supporting the long-term safety and efficacy of asciminib in this population., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
15. Payer and provider solutions to utilization management challenges in the management of rare hematologic cancers.
- Author
-
Jabbour EJ, Bobolts LR, Spinks TE, Geyer MB, Ebot-Tar VT, and Haumschild R
- Subjects
- Humans, Rare Diseases diagnosis, Rare Diseases therapy, Hematologic Neoplasms diagnosis, Hematologic Neoplasms therapy
- Abstract
Patients with rare diseases such as Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL), a hematologic malignancy affecting approximately 1500 new patients per year, experience barriers to care involving both clinical and administrative factors. Optimal patient outcomes depend on timely identification, diagnosis of disease, and treatment initiation. For patients living with Ph+ ALL, the process can be delayed by limited treatment options approved by the US Food and Drug Administration and administrative hurdles that often delay treatment initiation. An overhaul of utilization management processes, such as the requirement for prior authorization (PA) for treatment, are needed to ensure patients have access to appropriate treatments in a timely manner. An AJMC Roundtable in November 2022 brought together a panel of payers and providers to discuss the challenges and shortcomings of current PA processes and to present ideas for potential solutions for improving them. Panelists at the roundtable discussed approaches including the use of guideline-concordant electronic PAs and other digital solutions, expedited approval pathways for use in specific conditions, use of real-world evidence in decision-making, issuance of PA "Gold Cards" to select providers, and a shift to value-based care agreements. Roundtable attendees agreed that, regardless of the strategy for PA-process improvement, there is a need for improved communication between providers and payers to ensure that the decision-making system meets the essential need for timely patient access to optimal care. This article reviews utilization management and guideline-concordant care through the lens of rare diseases and then presents solutions to utilization.
- Published
- 2023
- Full Text
- View/download PDF
16. Molecular predictors of immunophenotypic measurable residual disease clearance in acute myeloid leukemia.
- Author
-
Stahl M, Derkach A, Farnoud N, Bewersdorf JP, Robinson T, Famulare C, Cho C, Devlin S, Menghrajani K, Patel MA, Cai SF, Miles LA, Bowman RL, Geyer MB, Dunbar A, Epstein-Peterson ZD, McGovern E, Schulman J, Glass JL, Taylor J, Viny AD, Stein EM, Getta B, Arcila ME, Gao Q, Barker J, Shaffer BC, Papadopoulos EB, Gyurkocza B, Perales MA, Abdel-Wahab O, Levine RL, Giralt SA, Zhang Y, Xiao W, Pai N, Papaemmanuil E, Tallman MS, Roshal M, and Goldberg AD
- Subjects
- Humans, Prognosis, Stem Cell Transplantation, Remission Induction, Transplantation, Homologous, Neoplasm, Residual genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Hematopoietic Stem Cell Transplantation
- Abstract
Measurable residual disease (MRD) is a powerful prognostic factor in acute myeloid leukemia (AML). However, pre-treatment molecular predictors of immunophenotypic MRD clearance remain unclear. We analyzed a dataset of 211 patients with pre-treatment next-generation sequencing who received induction chemotherapy and had MRD assessed by serial immunophenotypic monitoring after induction, subsequent therapy, and allogeneic stem cell transplant (allo-SCT). Induction chemotherapy led to MRD- remission, MRD+ remission, and persistent disease in 35%, 27%, and 38% of patients, respectively. With subsequent therapy, 34% of patients with MRD+ and 26% of patients with persistent disease converted to MRD-. Mutations in CEBPA, NRAS, KRAS, and NPM1 predicted high rates of MRD- remission, while mutations in TP53, SF3B1, ASXL1, and RUNX1 and karyotypic abnormalities including inv (3), monosomy 5 or 7 predicted low rates of MRD- remission. Patients with fewer individual clones were more likely to achieve MRD- remission. Among 132 patients who underwent allo-SCT, outcomes were favorable whether patients achieved early MRD- after induction or later MRD- after subsequent therapy prior to allo-SCT. As MRD conversion with chemotherapy prior to allo-SCT is rarely achieved in patients with specific baseline mutational patterns and high clone numbers, upfront inclusion of these patients into clinical trials should be considered., (© 2022 Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
17. Outcomes of relapsed B-cell acute lymphoblastic leukemia after sequential treatment with blinatumomab and inotuzumab.
- Author
-
Wudhikarn K, King AC, Geyer MB, Roshal M, Bernal Y, Gyurkocza B, Perales MA, and Park JH
- Subjects
- Adult, Antigens, CD19, Humans, Inotuzumab Ozogamicin, Recurrence, Antibodies, Bispecific adverse effects, Burkitt Lymphoma, Lymphoma, B-Cell, Precursor Cell Lymphoblastic Leukemia-Lymphoma
- Abstract
Novel monoclonal antibody (mAb)-based therapies targeting CD19 and CD22 (blinatumomab and inotuzumab) have shown high rates of complete remission (CR) and been used as a bridging treatment to potentially curative allogeneic hematopoietic stem cell transplantation (alloHSCT) in adults with relapsed or refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). However, limited data exist on the outcome of patients resistant to both mAbs as well as responses to each agent when progressed after the alternate antigen-targeted mAb. Herein, we report outcomes of 29 patients with R/R B-ALL previously treated with both blinatumomab and inotuzumab. Twenty-five patients (86.2%) received blinatumomab as first mAb (mAb1), and CD19-negative/dim relapses were observed in 44% of the patients. Inotuzumab induced CR in 68% of the patients for post-blinatumomab relapse regardless of CD19 expression status. The median time between mAb1 and mAb2 was 99 days. Twelve (63.2%) of 19 patients who achieved remission after mAb2 underwent alloHSCT. The median time from mAb2 to alloHSCT was 37.5 days. Acute graft-versus-host disease and nonrelapse mortality were observed in 58.3% (grade 3 or higher, 25%) and 41.7%, respectively. With a median follow-up of 16.8 months after mAb2, 19 patients (65.5%) relapsed, and 21 patients (72.4%) have died. Overall survival was not different between alloHSCT and non-alloHSCT patients. In conclusion, patients with B-ALL who relapsed after blinatumomab could be successfully rescued by inotuzumab as a bridge to alloHSCT but represent an ultra-high-risk group with poor overall survival. Further studies, including novel consolidation and treatment sequence, may improve outcomes of these patients., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
18. A phase 2 biomarker-driven study of ruxolitinib demonstrates effectiveness of JAK/STAT targeting in T-cell lymphomas.
- Author
-
Moskowitz AJ, Ghione P, Jacobsen E, Ruan J, Schatz JH, Noor S, Myskowski P, Vardhana S, Ganesan N, Hancock H, Davey T, Perez L, Ryu S, Santarosa A, Dowd J, Obadi O, Pomerantz L, Yi N, Sohail S, Galasso N, Neuman R, Liotta B, Blouin W, Baik J, Geyer MB, Noy A, Straus D, Kumar P, Dogan A, Hollmann T, Drill E, Rademaker J, Schoder H, Inghirami G, Weinstock DM, and Horwitz SM
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Janus Kinases antagonists & inhibitors, Lymphoma, T-Cell, Peripheral metabolism, Male, Middle Aged, Molecular Targeted Therapy, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local metabolism, Treatment Outcome, Young Adult, Janus Kinases metabolism, Lymphoma, T-Cell, Peripheral drug therapy, Nitriles therapeutic use, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use, Pyrimidines therapeutic use, STAT Transcription Factors metabolism
- Abstract
Signaling through JAK1 and/or JAK2 is common among tumor and nontumor cells within peripheral T-cell lymphoma (PTCL). No oral therapies are approved for PTCL, and better treatments for relapsed/refractory disease are urgently needed. We conducted a phase 2 study of the JAK1/2 inhibitor ruxolitinib for patients with relapsed/refractory PTCL (n = 45) or mycosis fungoides (MF) (n = 7). Patients enrolled onto 1 of 3 biomarker-defined cohorts: (1) activating JAK and/or STAT mutations, (2) ≥30% pSTAT3 expression among tumor cells by immunohistochemistry, or (3) neither or insufficient tissue to assess. Patients received ruxolitinib 20 mg PO twice daily until progression and were assessed for response after cycles 2 and 5 and every 3 cycles thereafter. The primary endpoint was clinical benefit rate (CBR), defined as the combination of complete response, partial response (PR), and stable disease lasting at least 6 months. Only 1 of 7 patients with MF had CBR (ongoing PR > 18 months). CBR among the PTCL cases (n = 45) in cohorts 1, 2, and 3 were 53%, 45%, and 13% (cohorts 1 & 2 vs 3, P = .02), respectively. Eight patients had CBR > 12 months (5 ongoing), including 4 of 5 patients with T-cell large granular lymphocytic leukemia. In an exploratory analysis using multiplex immunofluorescence, expression of phosphorylated S6, a marker of PI3 kinase or mitogen-activated protein kinase activation, in <25% of tumor cells was associated with response to ruxolitinib (P = .05). Our findings indicate that ruxolitinib is active across various PTCL subtypes and support a precision therapy approach to JAK/STAT inhibition in patients with PTCL. This trial was registered at www.clincialtrials.gov as #NCT02974647., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
19. Interventions and outcomes of adult patients with B-ALL progressing after CD19 chimeric antigen receptor T-cell therapy.
- Author
-
Wudhikarn K, Flynn JR, Rivière I, Gönen M, Wang X, Senechal B, Curran KJ, Roshal M, Maslak PG, Geyer MB, Halton EF, Diamonte C, Davila ML, Sadelain M, Brentjens RJ, and Park JH
- Subjects
- Adult, Aged, Disease-Free Survival, Female, Humans, Male, Middle Aged, Survival Rate, Antibodies, Bispecific administration & dosage, Immunotherapy, Adoptive, Inotuzumab Ozogamicin administration & dosage, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma mortality, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma therapy, Salvage Therapy
- Abstract
CD19-targeted chimeric antigen receptor (CAR) T-cell therapy has become a breakthrough treatment of patients with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). However, despite the high initial response rate, the majority of adult patients with B-ALL progress after CD19 CAR T-cell therapy. Data on the natural history, management, and outcome of adult B-ALL progressing after CD19 CAR T cells have not been described in detail. Herein, we report comprehensive data of 38 adult patients with B-ALL who progressed after CD19 CAR T therapy at our institution. The median time to progression after CAR T-cell therapy was 5.5 months. Median survival after post-CAR T progression was 7.5 months. A high disease burden at the time of CAR T-cell infusion was significantly associated with risk of post-CAR T progression. Thirty patients (79%) received salvage treatment of post-CAR T disease progression, and 13 patients (43%) achieved complete remission (CR), but remission duration was short. Notably, 7 (58.3%) of 12 patients achieved CR after blinatumomab and/or inotuzumab administered following post-CAR T failure. Multivariate analysis revealed that a longer remission duration from CAR T cells was associated with superior survival after progression following CAR T-cell therapy. In summary, overall prognosis of adult B-ALL patients progressing after CD19 CAR T cells was poor, although a subset of patients achieved sustained remissions to salvage treatments, including blinatumomab, inotuzumab, and reinfusion of CAR T cells. Novel therapeutic strategies are needed to reduce risk of progression after CAR T-cell therapy and improve outcomes of these patients., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
20. Neutropenia in adult acute myeloid leukemia patients represents a powerful risk factor for COVID-19 related mortality.
- Author
-
Stahl M, Narendra V, Jee J, Derkach A, Maloy M, Geyer MB, Mato AR, Roeker LE, Tallman MS, Shah GL, Daniyan A, and Goldberg AD
- Subjects
- Adult, COVID-19 Testing, Humans, Risk Factors, SARS-CoV-2, COVID-19, Leukemia, Myeloid, Acute complications, Leukemia, Myeloid, Acute epidemiology, Leukemia, Myeloid, Acute therapy, Neutropenia epidemiology, Neutropenia etiology
- Abstract
Patients with hematological malignancies are at risk for poor outcomes when diagnosed with coronavirus disease 2019 (COVID-19). It remains unclear whether cytopenias and specific leukemia subtypes play a role in the clinical course of COVID-19 infection. Here, we report outcomes and their clinical/laboratory predictors for 65 patients with acute and chronic leukemias diagnosed with COVID-19 between 8 March 2020 and 19 May 2020 at Memorial Sloan Kettering Cancer Center in New York City. Most patients had CLL (38%) or AML (26%). A total of 14 (22%) patients required high flow nasal cannula or were intubated for mechanical ventilation and 11 patients (17%) died. A diagnosis of AML (OR 4.7, p =.028), active treatment within the last 3 months (OR 5.22, p =.047), neutropenia within seven days prior and up to 28 days after SARS-CoV-2 diagnosis (11.75, p =.001) and ≥3 comorbidities (OR 6.55, p =.019) were associated with increased odds of death.
- Published
- 2021
- Full Text
- View/download PDF
21. Pediatric-inspired chemotherapy incorporating pegaspargase is safe and results in high rates of minimal residual disease negativity in adults up to age 60 with Philadelphia chromosome-negative acute lymphoblastic leukemia.
- Author
-
Geyer MB, Ritchie EK, Rao AV, Vemuri S, Flynn J, Hsu M, Devlin SM, Roshal M, Gao Q, Shukla M, Salcedo JM, Maslak P, Tallman MS, Douer D, and Park JH
- Subjects
- Adult, Antineoplastic Combined Chemotherapy Protocols adverse effects, Child, Humans, Middle Aged, Neoplasm, Residual, Philadelphia Chromosome, Polyethylene Glycols adverse effects, Young Adult, Asparaginase adverse effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Administration of pediatric-inspired chemotherapy to adults up to age 60 with acute lymphoblastic leukemia (ALL) is challenging in part due to toxicities of asparaginase as well as myelosuppression. We conducted a multicenter phase II clinical trial (NCT01920737) investigating a pediatric-inspired regimen, based on the augmented arm of the Children's Cancer Group 1882 protocol, incorporating 6 doses of pegaspargase 2000 IU/m2, rationally synchronized to avoid overlapping toxicity with other agents. We treated 39 adults ages 20-60 years (median, 38 years) with newly-diagnosed ALL (n=31) or lymphoblastic lymphoma (n=8). Grade 3-4 hyperbilirubinemia occurred frequently and at higher rates in patients 40-60 (n=18) vs 18-39 (n=21) years (44 vs 10%, p=0.025). However, 8/9 patients re-challenged with pegaspargase did not experience recurrent grade 3-4 hyperbilirubinemia. Grade 3-4 hypertriglyceridemia and hypofibrinogenemia were common (each 59%). Asparaginase activity at 7-days post-infusion reflected levels associated with adequate asparagine depletion, even among those with antibodies to pegaspargase. Complete response (CR)/CR with incomplete hematologic recovery was observed post-induction in 38/39 (97%) patients. Among patients with ALL, rates of MRD negativity by multiparameter flow cytometry were 33% and 83% following Induction Phase I and Phase II, respectively. Event-free and overall survival at 3 years (67.8 and 76.4%) compare favorably to outcomes observed in other series. These results demonstrate pegaspargase can be administered in the context of intensive multi-agent chemotherapy to adults age ≤60 with manageable toxicity. This regimen may serve as an effective backbone into which novel agents may be incorporated in future frontline studies.
- Published
- 2021
- Full Text
- View/download PDF
22. Depletion of high-content CD14 + cells from apheresis products is critical for successful transduction and expansion of CAR T cells during large-scale cGMP manufacturing.
- Author
-
Wang X, Borquez-Ojeda O, Stefanski J, Du F, Qu J, Chaudhari J, Thummar K, Zhu M, Shen LB, Hall M, Gautam P, Wang Y, Sénéchal B, Sikder D, Adusumilli PS, Brentjens RJ, Curran K, Geyer MB, Mailankhody S, O'Cearbhaill R, Park JH, Sauter C, Slovin S, Smith EL, and Rivière I
- Abstract
With the US Food and Drug Administration (FDA) approval of four CD19- and one BCMA-targeted chimeric antigen receptor (CAR) therapy for B cell malignancies, CAR T cell therapy has finally reached the status of a medicinal product. The successful manufacturing of autologous CAR T cell products is a key requirement for this promising treatment modality. By analyzing the composition of 214 apheresis products from 210 subjects across eight disease indications, we found that high CD14
+ cell content poses a challenge for manufacturing CAR T cells, especially in patients with non-Hodgkin's lymphoma and multiple myeloma caused by the non-specific phagocytosis of the magnetic beads used to activate CD3+ T cells. We demonstrated that monocyte depletion via rapid plastic surface adhesion significantly reduces the CD14+ monocyte content in the apheresis products and simultaneously boosts the CD3+ content. We established a 40% CD14+ threshold for the stratification of apheresis products across nine clinical trials and demonstrated the effectiveness of this procedure by comparing manufacturing runs in two phase 1 clinical trials. Our study suggests that CD14+ content should be monitored in apheresis products, and that the manufacturing of CAR T cells should incorporate a step that lessens the CD14+ cell content in apheresis products containing more than 40% to maximize the production success., Competing Interests: P.S.A. has received research funding from ATARA Biotherapeutics; has served on the Scientific Advisory Board or as consultant to ATARA Biotherapeutics, Bayer, Carisma Therapeutics, Imugene, and Takeda Therapeutics; and has patents, royalties, and intellectual property on mesothelin-targeted CARs and other T cell therapies, method for detection of cancer cells using virus, and pending patent applications on T cell therapies. E.L.S. has patents, royalties, and intellectual property on BCMA-targeted CARs and serves as consultant for BMS. I.R. has intellectual property rights from Juno Therapeutics., (© 2021.)- Published
- 2021
- Full Text
- View/download PDF
23. Tolerability and toxicity of pegaspargase in adults 40 years and older with acute lymphoblastic leukemia.
- Author
-
Daley RJ, Rajeeve S, Kabel CC, Pappacena JJ, Stump SE, Lavery JA, Tallman MS, Geyer MB, and Park JH
- Subjects
- Adult, Asparaginase adverse effects, Asparagine, Humans, Polyethylene Glycols adverse effects, Antineoplastic Agents therapeutic use, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Abstract
Pegaspargase is a modified version of asparaginase with prolonged asparagine depletion. It appears to be safe in adults <40 years old, but has a unique spectrum of toxicities, the risks of which appear to increase with age. The primary objective of this study was to evaluate pegaspargase tolerability and toxicity as assessed by evaluation of incidence and severity of adverse events. Secondary objectives included characterization of the reasons underlying pegaspargase discontinuation, when applicable. Grade 3/4 asparaginase-related toxicities with ≥10% incidence included: hyperbilirubinemia, hyperglycemia, hypertriglyceridemia, hypoalbuminemia, hypofibrinogenemia, and transaminitis. 63% of patients (38 of 60) received all intended doses of pegaspargase, with the most common reasons for discontinuation noted as hypersensitivity (12%), hyperbilirubinemia/transaminitis (8%), and hematopoietic transplantation in complete remission (10%). This study suggests that while hepatotoxicity and other known adverse effects are common, with careful monitoring, pegaspargase can safely be administered to adults with ALL age ≥40 years old.
- Published
- 2021
- Full Text
- View/download PDF
24. Hypofibrinogenemia and disseminated intravascular coagulation rarely complicate treatment-naïve acute lymphoblastic leukemia.
- Author
-
Gaulin C, Chan A, Derkach A, Park JH, Mantha S, Geyer MB, and Tallman MS
- Subjects
- Humans, Afibrinogenemia complications, Afibrinogenemia diagnosis, Disseminated Intravascular Coagulation diagnosis, Disseminated Intravascular Coagulation etiology, Precursor Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy
- Published
- 2020
- Full Text
- View/download PDF
25. Outcomes of COVID-19 in patients with CLL: a multicenter international experience.
- Author
-
Mato AR, Roeker LE, Lamanna N, Allan JN, Leslie L, Pagel JM, Patel K, Osterborg A, Wojenski D, Kamdar M, Huntington SF, Davids MS, Brown JR, Antic D, Jacobs R, Ahn IE, Pu J, Isaac KM, Barr PM, Ujjani CS, Geyer MB, Berman E, Zelenetz AD, Malakhov N, Furman RR, Koropsak M, Bailey N, Hanson L, Perini GF, Ma S, Ryan CE, Wiestner A, Portell CA, Shadman M, Chong EA, Brander DM, Sundaram S, Seddon AN, Seymour E, Patel M, Martinez-Calle N, Munir T, Walewska R, Broom A, Walter H, El-Sharkawi D, Parry H, Wilson MR, Patten PEM, Hernández-Rivas JÁ, Miras F, Fernández Escalada N, Ghione P, Nabhan C, Lebowitz S, Bhavsar E, López-Jiménez J, Naya D, Garcia-Marco JA, Skånland SS, Cordoba R, and Eyre TA
- Subjects
- Adult, Agammaglobulinaemia Tyrosine Kinase antagonists & inhibitors, Aged, Aged, 80 and over, Anti-Inflammatory Agents therapeutic use, Antiviral Agents therapeutic use, Betacoronavirus isolation & purification, COVID-19, Coronavirus Infections therapy, Female, Humans, Immunization, Passive, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Male, Middle Aged, Pandemics, Pneumonia, Viral therapy, Protein Kinase Inhibitors therapeutic use, SARS-CoV-2, Survival Analysis, Treatment Outcome, COVID-19 Serotherapy, Coronavirus Infections complications, Leukemia, Lymphocytic, Chronic, B-Cell complications, Pneumonia, Viral complications
- Abstract
Given advanced age, comorbidities, and immune dysfunction, chronic lymphocytic leukemia (CLL) patients may be at particularly high risk of infection and poor outcomes related to coronavirus disease 2019 (COVID-19). Robust analysis of outcomes for CLL patients, particularly examining effects of baseline characteristics and CLL-directed therapy, is critical to optimally manage CLL patients through this evolving pandemic. CLL patients diagnosed with symptomatic COVID-19 across 43 international centers (n = 198) were included. Hospital admission occurred in 90%. Median age at COVID-19 diagnosis was 70.5 years. Median Cumulative Illness Rating Scale score was 8 (range, 4-32). Thirty-nine percent were treatment naive ("watch and wait"), while 61% had received ≥1 CLL-directed therapy (median, 2; range, 1-8). Ninety patients (45%) were receiving active CLL therapy at COVID-19 diagnosis, most commonly Bruton tyrosine kinase inhibitors (BTKi's; n = 68/90 [76%]). At a median follow-up of 16 days, the overall case fatality rate was 33%, though 25% remain admitted. Watch-and-wait and treated cohorts had similar rates of admission (89% vs 90%), intensive care unit admission (35% vs 36%), intubation (33% vs 25%), and mortality (37% vs 32%). CLL-directed treatment with BTKi's at COVID-19 diagnosis did not impact survival (case fatality rate, 34% vs 35%), though the BTKi was held during the COVID-19 course for most patients. These data suggest that the subgroup of CLL patients admitted with COVID-19, regardless of disease phase or treatment status, are at high risk of death. Future epidemiologic studies are needed to assess severe acute respiratory syndrome coronavirus 2 infection risk, these data should be validated independently, and randomized studies of BTKi's in COVID-19 are needed to provide definitive evidence of benefit.
- Published
- 2020
- Full Text
- View/download PDF
26. Considerations for Use of Hematopoietic Growth Factors in Patients With Cancer Related to the COVID-19 Pandemic.
- Author
-
Griffiths EA, Alwan LM, Bachiashvili K, Brown A, Cool R, Curtin P, Geyer MB, Gojo I, Kallam A, Kidwai WZ, Kloth DD, Kraut EH, Lyman GH, Mukherjee S, Perez LE, Rosovsky RP, Roy V, Rugo HS, Vasu S, Wadleigh M, Westervelt P, and Becker PS
- Abstract
Hematopoietic growth factors, including erythrocyte stimulating agents (ESAs), granulocyte colony-stimulating factors, and thrombopoietin mimetics, can mitigate anemia, neutropenia, and thrombocytopenia resulting from chemotherapy for the treatment of cancer. In the context of pandemic SARS-CoV-2 infection, patients with cancer have been identified as a group at high risk of morbidity and mortality from this infection. Our subcommittee of the NCCN Hematopoietic Growth Factors Panel convened a voluntary group to review the potential value of expanded use of such growth factors in the current high-risk environment. Although recommendations are available on the NCCN website in the COVID-19 Resources Section (https://www.nccn.org/covid-19/), these suggestions are provided without substantial context or reference. Herein we review the rationale and data underlying the suggested alterations to the use of hematopoietic growth factors for patients with cancer in the COVID-19 era.
- Published
- 2020
- Full Text
- View/download PDF
27. Allogeneic stem cell transplantation for chronic lymphocytic leukemia in the era of novel agents.
- Author
-
Roeker LE, Dreger P, Brown JR, Lahoud OB, Eyre TA, Brander DM, Skarbnik A, Coombs CC, Kim HT, Davids M, Manchini ST, George G, Shah N, Voorhees TJ, Orchard KH, Walter HS, Arumainathan AK, Sitlinger A, Park JH, Geyer MB, Zelenetz AD, Sauter CS, Giralt SA, Perales MA, and Mato AR
- Subjects
- Humans, Retrospective Studies, Graft vs Host Disease etiology, Hematopoietic Stem Cell Transplantation, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Lymphoma, Follicular
- Abstract
Although novel agents (NAs) have improved outcomes for patients with chronic lymphocytic leukemia (CLL), a subset will progress through all available NAs. Understanding outcomes for potentially curative modalities including allogeneic hematopoietic stem cell transplantation (alloHCT) following NA therapy is critical while devising treatment sequences aimed at long-term disease control. In this multicenter, retrospective cohort study, we examined 65 patients with CLL who underwent alloHCT following exposure to ≥1 NA, including baseline disease and transplant characteristics, treatment preceding alloHCT, transplant outcomes, treatment following alloHCT, and survival outcomes. Univariable and multivariable analyses evaluated associations between pre-alloHCT factors and progression-free survival (PFS). Twenty-four-month PFS, overall survival (OS), nonrelapse mortality, and relapse incidence were 63%, 81%, 13%, and 27% among patients transplanted for CLL. Day +100 cumulative incidence of grade III-IV acute graft-vs-host disease (GVHD) was 24%; moderate-severe GVHD developed in 27%. Poor-risk disease characteristics, prior NA exposure, complete vs partial remission, and transplant characteristics were not independently associated with PFS. Hematopoietic cell transplantation-specific comorbidity index independently predicts PFS. PFS and OS were not impacted by having received NAs vs both NAs and chemoimmunotherapy, 1 vs ≥2 NAs, or ibrutinib vs venetoclax as the line of therapy immediately pre-alloHCT. AlloHCT remains a viable long-term disease control strategy that overcomes adverse CLL characteristics. Prior NAs do not appear to impact the safety of alloHCT, and survival outcomes are similar regardless of number of NAs received, prior chemoimmunotherapy exposure, or NA immediately preceding alloHCT. Decisions about proceeding to alloHCT should consider comorbidities and anticipated response to remaining therapeutic options., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
28. FGFR1 Rearrangement Guides Diagnosis and Treatment of a Trilineage B, T, and Myeloid Mixed Phenotype Acute Leukemia.
- Author
-
Liu Y, Mi X, Gadde R, Gao Q, Xiao W, Zhang Y, Benayed R, Arcila M, Dogan A, Geyer MB, and Roshal M
- Abstract
Competing Interests: The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center. Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments). Wenbin XiaoResearch Funding: Stemline Therapeutics (Inst)Maria ArcilaHonoraria: Invivoscribe, Biocartis Consulting or Advisory Role: AstraZeneca Travel, Accommodations, Expenses: AstraZeneca, Invivoscribe, Raindance TechnologiesAhmet DoganConsulting or Advisory Role: Seattle Genetics, Roche, Takeda, EUSA Pharma, Abbvie Research Funding: GenentechMark B. GeyerResearch Funding: Amgen (Inst)Mikhail RoshalHonoraria: Celgene Consulting or Advisory Role: Agios, Auron Research Funding: Agios, Roche Research Funding: Boehringer Ingelheim, Boehringer Ingelheim No other potential conflicts of interest were reported.
- Published
- 2020
- Full Text
- View/download PDF
29. Inotuzumab ozogamicin as chemotherapy-sparing salvage in a 67-year-old man with primary refractory B-cell acute lymphoblastic leukemia with high-risk genomic features.
- Author
-
Yang X, Sen F, and Geyer MB
- Abstract
Older adults with acute lymphoblastic leukemia (ALL) continue to have a poor prognosis, in part due to greater chemotherapy-related toxicities. We herein report a 67-year-old man with Philadelphia chromosome (Ph)-negative B-cell ALL, who exhibited refractoriness to 3 different regimens of induction chemotherapy and experienced multiple complications including intracranial bleeding and respiratory failure, who achieved minimal residual disease (MRD)-negative complete response (CR) after a single cycle of inotuzumab ozogamicin (IO). His ALL was characterized by several high-risk mutations, which may have contributed to chemotherapy-refractory disease. Our case supports incorporating IO into front-line induction regimens for older adults with high-risk B-cell ALL., (© 2019 The Authors.)
- Published
- 2019
- Full Text
- View/download PDF
30. Loss of plasmacytoid dendritic cell differentiation is highly predictive for post-induction measurable residual disease and inferior outcomes in acute myeloid leukemia.
- Author
-
Xiao W, Goldberg AD, Famulare CA, Devlin SM, Nguyen NT, Sim S, Kabel CC, Patel MA, McGovern EM, Patel A, Schulman J, Dunbar AJ, Epstein-Peterson ZD, Menghrajani KN, Getta BM, Cai SF, Geyer MB, Glass JL, Taylor J, Viny AD, Levine RL, Zhang Y, Giralt SA, Klimek V, Tallman MS, and Roshal M
- Subjects
- Adult, Aged, Case-Control Studies, Combined Modality Therapy, Female, Follow-Up Studies, Humans, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute therapy, Male, Middle Aged, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local therapy, Neoplasm, Residual pathology, Neoplasm, Residual therapy, Prognosis, Retrospective Studies, Survival Rate, Dendritic Cells pathology, Leukemia, Myeloid, Acute mortality, Neoplasm Recurrence, Local mortality, Neoplasm, Residual mortality
- Abstract
Measurable residual disease is associated with inferior outcomes in patients with acute myeloid leukemia (AML). Measurable residual disease monitoring enhances risk stratification and may guide therapeutic intervention. The European LeukemiaNet working party recently came to a consensus recommendation incorporating leukemia associated immunophenotype-based different from normal approach by multi-color flow cytometry for measurable residual disease evaluation. However, the analytical approach is highly expertise-dependent and difficult to standardize. Here we demonstrate that loss of plasmacytoid dendritic cell differentiation after 7+3 induction in AML is highly specific for measurable residual disease positivity (specificity 97.4%) in a uniformly treated patient cohort. Moreover, loss of plasmacytoid dendritic cell differentiation as determined by a blast-to-plasmacytoid dendritic cell ratio >10 was strongly associated with inferior overall and relapse-free survival (RFS) [Hazard ratio 2.79, 95% confidence interval (95%CI): 0.98-7.97; P =0.077) and 3.83 (95%CI: 1.51-9.74; P =0.007), respectively), which is similar in magnitude to measurable residual disease positivity. Importantly, measurable residual disease positive patients who reconstituted plasmacytoid dendritic cell differentiation (blast/ plasmacytoid dendritic cell ratio <10) showed a higher rate of measurable residual disease clearance at later pre-transplant time points compared to patients with loss of plasmacytoid dendritic cell differentiation (blast/ plasmacytoid dendritic cell ratio <10) (6 of 12, 50% vs 2 of 18, 11%; P =0.03). Furthermore pre-transplant plasmacytoid dendritic cell recovery was associated with superior outcome in measurable residual disease positive patients. Our study provides a novel, simple, broadly applicable, and quantitative multi-color flow cytometry approach to risk stratification in AML., (Copyright© 2019 Ferrata Storti Foundation.)
- Published
- 2019
- Full Text
- View/download PDF
31. BCMA-Targeted CAR T-cell Therapy plus Radiotherapy for the Treatment of Refractory Myeloma Reveals Potential Synergy.
- Author
-
Smith EL, Mailankody S, Staehr M, Wang X, Senechal B, Purdon TJ, Daniyan AF, Geyer MB, Goldberg AD, Mead E, Santomasso BD, Landa J, Rimner A, Riviere I, Landgren O, and Brentjens RJ
- Subjects
- Combined Modality Therapy, Female, Humans, Middle Aged, Multiple Myeloma immunology, Remission Induction, B-Cell Maturation Antigen immunology, Drug Resistance, Neoplasm, Immunotherapy, Adoptive methods, Multiple Myeloma therapy, Radiotherapy methods, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology
- Abstract
We present a case of a patient with multiply relapsed, refractory myeloma whose clinical course showed evidence of a synergistic abscopal-like response to chimeric antigen receptor (CAR) T-cell therapy and localized radiotherapy (XRT). Shortly after receiving B-cell maturation antigen (BCMA)-targeted CAR T-cell therapy, the patient required urgent high-dose steroids and XRT for spinal cord compression. Despite the steroids, the patient had a durable systemic response that could not be attributed to XRT alone. Post-XRT findings included a second wave of fever and increased CRP and IL6, beginning 21 days after CAR T cells, which is late for cytokine-release syndrome from CAR T-cell therapy alone on this trial. Given this response, which resembled cytokine-release syndrome, immediately following XRT, we investigated changes in the patient's T-cell receptor (TCR) repertoire over 10 serial time points. Comparing T-cell diversity via Morisita's overlap indices ( C
D ), we discovered that, although the diversity was initially stable after CAR T-cell therapy compared with baseline ( CD = 0.89-0.97, baseline vs. 4 time points after CAR T cells), T-cell diversity changed after the conclusion of XRT, with >30% newly expanded TCRs ( CD = 0.56-0.69, baseline vs. 4 time points after XRT). These findings suggest potential synergy between radiation and CAR T-cell therapies resulting in an abscopal-like response., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
32. Safety and tolerability of conditioning chemotherapy followed by CD19-targeted CAR T cells for relapsed/refractory CLL.
- Author
-
Geyer MB, Rivière I, Sénéchal B, Wang X, Wang Y, Purdon TJ, Hsu M, Devlin SM, Palomba ML, Halton E, Bernal Y, van Leeuwen DG, Sadelain M, Park JH, and Brentjens RJ
- Subjects
- Adenine analogs & derivatives, Adult, Aged, Antigens, CD19 immunology, Antineoplastic Combined Chemotherapy Protocols adverse effects, Chemotherapy, Adjuvant adverse effects, Chemotherapy, Adjuvant methods, Cytokine Release Syndrome immunology, Disease-Free Survival, Drug Resistance, Neoplasm, Female, Follow-Up Studies, Humans, Immunotherapy, Adoptive adverse effects, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell mortality, Lymphoma, B-Cell immunology, Lymphoma, B-Cell mortality, Male, Middle Aged, Neoadjuvant Therapy adverse effects, Neoadjuvant Therapy methods, Neoplasm Recurrence, Local immunology, Piperidines, Pyrazoles administration & dosage, Pyrazoles adverse effects, Pyrimidines administration & dosage, Pyrimidines adverse effects, Receptors, Chimeric Antigen immunology, Transplantation Conditioning adverse effects, Transplantation, Autologous adverse effects, Transplantation, Autologous methods, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Cytokine Release Syndrome epidemiology, Immunotherapy, Adoptive methods, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Lymphoma, B-Cell therapy, Neoplasm Recurrence, Local therapy, Transplantation Conditioning methods
- Abstract
Background: Subgroups of patients with relapsed or refractory (R/R) chronic lymphocytic leukemia (CLL) exhibit suboptimal outcomes after standard therapies, including oral kinase inhibitors. We and others have previously reported on safety and efficacy of autologous CD19-targeted CAR T-cells for these patients; here we report safety and long-term follow-up of CAR T-cell therapy with or without conditioning chemotherapy for patients with R/R CLL and indolent B-cell non-Hodgkin lymphoma (B-NHL)., Methods: We conducted a phase 1 clinical trial investigating CD19-targeted CAR T-cells incorporating a CD28 costimulatory domain (19-28z). Seventeen of 20 patients received conditioning chemotherapy prior to CAR T-cell infusion. Five patients with CLL received ibrutinib at the time of autologous T-cell collection and/or CAR T-cell administration., Results: This analysis included 16 patients with R/R CLL and 4 patients with R/R indolent B-NHL. Cytokine release syndrome (CRS) was observed in all 20 patients but grades 3 and 4 CRS and neurological events were uncommon (10% for each). Ex vivo expansion of T-cells and proportions of CD4+/CD8+ CAR T-cells with CD62L+CD127+ immunophenotype were significantly greater in patients on ibrutinib at leukapheresis. Three of 12 evaluable CLL patients receiving conditioning chemotherapy achieved CR (two had minimal residual disease-negative CR). All patients achieving CR remained progression-free at median follow-up of 53 months., Conclusion: Conditioning chemotherapy and 19-28z CAR T-cells were acceptably tolerated across investigated dose levels in heavily pretreated patients with R/R CLL and indolent B-NHL, and a subgroup of patients achieved durable CR. Ibrutinib therapy may modulate autologous T-cell phenotype., Trial Registration: ClinicalTrials.gov NCT00466531., Funding: Juno Therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
33. Blinatumomab administered concurrently with oral tyrosine kinase inhibitor therapy is a well-tolerated consolidation strategy and eradicates measurable residual disease in adults with Philadelphia chromosome positive acute lymphoblastic leukemia.
- Author
-
King AC, Pappacena JJ, Tallman MS, Park JH, and Geyer MB
- Subjects
- Administration, Oral, Adult, Aged, Antibodies, Bispecific adverse effects, Consolidation Chemotherapy adverse effects, Female, Humans, Male, Middle Aged, Neoplasm, Residual, Philadelphia Chromosome, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Protein Kinase Inhibitors adverse effects, Protein-Tyrosine Kinases antagonists & inhibitors, Retrospective Studies, Treatment Outcome, Antibodies, Bispecific administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Protein Kinase Inhibitors administration & dosage
- Abstract
Incorporation of ABL-targeted oral tyrosine kinase inhibitors (TKIs) into frontline therapeutic regimens has improved outcomes for adults with Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph + ALL). However, patients with persistent minimal residual disease (MRD) exhibit increased risk of relapse. Combining consolidative chemotherapy with TKIs may increase rates of infectious complications, organ toxicity, hospitalization, and non-relapse mortality. Blinatumomab has demonstrated single-agent activity in patients with relapsed B-ALL or persistent MRD, including Ph + B-ALL. We have used blinatumomab concomitantly with commercially available TKIs as consolidative therapy to spare toxicities of conventional chemotherapy. We evaluated 11 adults with previously treated Ph + B-ALL who received blinatumomab concurrent with TKI (ponatinib, n = 5; dasatinib, n = 4; nilotinib, n = 1; imatinib, n = 1) to eradicate MRD or sustain MRD-negativity. Eight of 9 patients with MRD achieved BCR-ABL1 negativity (complete molecular response, CMR) after a median of one cycle; 2/2 patients without measurable disease durably maintained CMR. Cytokine release syndrome (all grade 1-2) was observed in 3/11 patients; one patient experienced transient grade 1 neurologic toxicity. Transient grade 2 transaminitis was observed in 6/11 patients, including 4/5 recipients of blinatumomab + ponatinib. This small series suggests blinatumomab + TKI is a safe and effective consolidation strategy for patients with Ph + ALL to achieve or maintain CMR., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
34. BRAF in the cross-hairs.
- Author
-
Geyer MB, Abdel-Wahab O, and Tallman MS
- Subjects
- Animals, Antineoplastic Agents adverse effects, Humans, Leukemia, Hairy Cell genetics, MAP Kinase Signaling System drug effects, Point Mutation drug effects, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Proto-Oncogene Proteins B-raf genetics, Signal Transduction drug effects, Vemurafenib adverse effects, Antineoplastic Agents therapeutic use, Leukemia, Hairy Cell drug therapy, Leukemia, Hairy Cell metabolism, Proto-Oncogene Proteins B-raf metabolism, Vemurafenib therapeutic use
- Abstract
Introduction: Hairy cell leukemia (HCL) is a rare, chronic B-cell lymphoproliferative disorder characterized by distinctive morphologic features and an indolent clinical course. The discovery of a recurrent activating mutation in BRAF (BRAF V600E) as a disease-defining genetic event in HCL has substantial diagnostic and therapeutic implications. Areas covered: Herein the authors review the role of BRAF V600E and RAF-MEK-ERK signaling in the pathogenesis of HCL, anecdotal clinical reports of BRAF inhibitor monotherapy in management of relapsed or refractory HCL, larger phase 2 trials investigating efficacy of BRAF inhibitor therapy for HCL, adverse effects commonly associated with BRAF inhibitor therapy, including cutaneous toxicity, and mechanisms of therapeutic resistance. Expert opinion: Ongoing and planned studies will help to optimize the use of BRAF inhibitor therapy for HCL by determining the efficacy of BRAF inhibition in combination with other antigen targeted or molecularly targeted therapies, and more broadly, to determine how hematologists can best utilize and sequence emerging diagnostic and therapeutic modalities in the care of patients with newly diagnosed and relapsed or refractory HCL.
- Published
- 2019
- Full Text
- View/download PDF
35. First CAR to Pass the Road Test: Tisagenlecleucel's Drive to FDA Approval.
- Author
-
Geyer MB
- Subjects
- B-Lymphocytes, Humans, Immunotherapy, Adoptive, Precursor Cell Lymphoblastic Leukemia-Lymphoma, Receptors, Antigen, T-Cell
- Abstract
In August 2017, the FDA took historic action in granting the first approval of gene therapy to tisagenlecleucel. This landmark step brought CAR T-cell therapy to the commercial space and heralded a new era in managing refractory B-cell malignancies and FDA oversight of gene-modified therapies.See related article by O'Leary et al., p. 1142., (©2018 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
36. The safety and efficacy of clofarabine in combination with high-dose cytarabine and total body irradiation myeloablative conditioning and allogeneic stem cell transplantation in children, adolescents, and young adults (CAYA) with poor-risk acute leukemia.
- Author
-
Hochberg J, Zahler S, Geyer MB, Chen N, Krajewski J, Harrison L, Militano O, Ozkaynak MF, Cheerva AC, Talano J, Moore TB, Gillio AP, Walters MC, Baxter-Lowe LA, Hamby C, and Cairo MS
- Subjects
- Acute Disease, Adolescent, Child, Child, Preschool, Clofarabine therapeutic use, Cytarabine therapeutic use, Female, Hematopoietic Stem Cell Transplantation adverse effects, Hematopoietic Stem Cell Transplantation mortality, Humans, Infant, Leukemia complications, Leukemia mortality, Leukemia, Myeloid, Acute therapy, Male, Myeloablative Agonists adverse effects, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Transplantation Conditioning adverse effects, Transplantation Conditioning mortality, Transplantation, Homologous, Treatment Outcome, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Hematopoietic Stem Cell Transplantation methods, Leukemia therapy, Myeloablative Agonists therapeutic use, Transplantation Conditioning methods, Whole-Body Irradiation methods
- Abstract
Acute leukemias in children with CR3, refractory relapse, or induction failure (IF) have a poor prognosis. Clofarabine has single agent activity in relapsed leukemia and synergy with cytarabine. We sought to determine the safety and overall survival in a Phase I/II trial of conditioning with clofarabine (doses 40 - 52 mg/m
2 ), cytarabine 1000 mg/m2 , and 1200 cGy TBI followed by alloSCT in children, adolescents, and young adults with poor-risk leukemia. Thirty-seven patients; Age 12 years (1-22 years); ALL/AML: 34:3 (18 IF, 10 CR3, 13 refractory relapse); 15 related, 22 unrelated donors. Probabilities of neutrophil, platelet engraftment, acute GvHD, and chronic GvHD were 94%, 84%, 49%, and 30%, respectively. Probability of day 100 TRM was 8.1%. 2-year EFS (event free survival) and OS (overall survival) were 38.6% (CI95 : 23-54%), and 41.3% (CI95 : 25-57%). Multivariate analysis demonstrated overt disease at time of transplant (relative risk (RR) 3.65, CI95 : 1.35-9.89, P = 0.011) and umbilical cord blood source (RR 2.17, CI95 : 1.33-4.15, P = 0.019) to be predictors of worse EFS/OS. This novel myeloablative conditioning regimen followed by alloSCT is safe and well tolerated in CAYA with very poor-risk ALL or AML. Further investigation in CAYA with better risk ALL and AML undergoing alloSCT is warranted.- Published
- 2019
- Full Text
- View/download PDF
37. Autologous CD19-Targeted CAR T Cells in Patients with Residual CLL following Initial Purine Analog-Based Therapy.
- Author
-
Geyer MB, Rivière I, Sénéchal B, Wang X, Wang Y, Purdon TJ, Hsu M, Devlin SM, Halton E, Lamanna N, Rademaker J, Sadelain M, Brentjens RJ, and Park JH
- Subjects
- Aged, Behavior Therapy, Cyclophosphamide therapeutic use, Female, Humans, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Male, Middle Aged, Neoplasm, Residual, Pentostatin therapeutic use, Rituximab therapeutic use, T-Lymphocytes immunology, Transplantation, Autologous adverse effects, Treatment Outcome, Antigens, CD19 metabolism, Cyclophosphamide administration & dosage, Immunotherapy, Adoptive adverse effects, Leukemia, Lymphocytic, Chronic, B-Cell therapy, T-Lymphocytes transplantation
- Abstract
Patients with residual chronic lymphocytic leukemia (CLL) following initial purine analog-based chemoimmunotherapy exhibit a shorter duration of response and may benefit from novel therapeutic strategies. We and others have previously described the safety and efficacy of autologous T cells modified to express anti-CD19 chimeric antigen receptors (CARs) in patients with relapsed or refractory B cell acute lymphoblastic leukemia and CLL. Here we report the use of CD19-targeted CAR T cells incorporating the intracellular signaling domain of CD28 (19-28z) as a consolidative therapy in 8 patients with residual CLL following first-line chemoimmunotherapy with pentostatin, cyclophosphamide, and rituximab. Outpatients received low-dose conditioning therapy with cyclophosphamide (600 mg/m
2 ), followed by escalating doses of 3 × 106 , 1 × 107 , or 3 × 107 19-28z CAR T cells/kg. An objective response was observed in 3 of 8 patients (38%), with a clinically complete response lasting more than 28 months observed in two patients. Self-limited fevers were observed post-CAR T cell infusion in 4 patients, contemporaneous with elevations in interleukin-6 (IL-6), IL-10, IL-2, and TGF-α. None developed severe cytokine release syndrome or neurotoxicity. CAR T cells were detectable post-infusion in 4 patients, with a longest observed persistence of 48 days by qPCR. Further strategies to enhance CAR T cell efficacy in CLL are under investigation., (Copyright © 2018. Published by Elsevier Inc.)- Published
- 2018
- Full Text
- View/download PDF
38. Concurrent therapy of chronic lymphocytic leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia utilizing CD19-targeted CAR T-cells.
- Author
-
Geyer MB, Manjunath SH, Evans AG, Park JH, Davila ML, Cutler CS, Wang X, Wang Y, Senechal B, Rivière I, Sadelain M, Liesveld JL, and Brentjens RJ
- Subjects
- Antigens, CD19 metabolism, Biomarkers, Biopsy, Combined Modality Therapy, Humans, Middle Aged, Precursor Cell Lymphoblastic Leukemia-Lymphoma diagnosis, Receptors, Antigen, T-Cell genetics, Receptors, Chimeric Antigen genetics, Treatment Outcome, Antigens, CD19 immunology, Immunotherapy, Adoptive methods, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Receptors, Antigen, T-Cell metabolism, Receptors, Chimeric Antigen metabolism
- Published
- 2018
- Full Text
- View/download PDF
39. Digging deeper in relapsed acute lymphoblastic leukemia: impact of MRD status on outcome in second remission.
- Author
-
Geyer MB and Tallman MS
- Subjects
- Humans, Remission Induction, Neoplasm, Residual, Precursor Cell Lymphoblastic Leukemia-Lymphoma
- Published
- 2018
- Full Text
- View/download PDF
40. Overall survival among older US adults with ALL remains low despite modest improvement since 1980: SEER analysis.
- Author
-
Geyer MB, Hsu M, Devlin SM, Tallman MS, Douer D, and Park JH
- Subjects
- Adult, Disease-Free Survival, Female, Humans, Male, Middle Aged, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Survival Rate, Databases, Factual, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality, SEER Program
- Published
- 2017
- Full Text
- View/download PDF
41. Review: Current clinical applications of chimeric antigen receptor (CAR) modified T cells.
- Author
-
Geyer MB and Brentjens RJ
- Subjects
- Animals, Humans, Neoplasms immunology, Neoplasms therapy, Clinical Trials as Topic, Immunotherapy, Adoptive methods, Receptors, Antigen, T-Cell immunology, T-Lymphocytes immunology
- Abstract
The past several years have been marked by extraordinary advances in clinical applications of immunotherapy. In particular, adoptive cellular therapy utilizing chimeric antigen receptor (CAR)-modified T cells targeted to CD19 has demonstrated substantial clinical efficacy in children and adults with relapsed or refractory B-cell acute lymphoblastic leukemia (B-ALL) and durable clinical benefit in a smaller subset of patients with relapsed or refractory chronic lymphocytic leukemia (CLL) or B-cell non-Hodgkin lymphoma (B-NHL). Early-phase clinical trials are currently assessing CAR T-cell safety and efficacy in additional malignancies. Here, we discuss clinical results from the largest series to date investigating CD19-targeted CAR T cells in B-ALL, CLL, and B-NHL, including discussion of differences in CAR T-cell design and production and treatment approach, as well as clinical efficacy, nature of severe cytokine release syndrome and neurologic toxicities, and CAR T-cell expansion and persistence. We additionally review the current and forthcoming use of CAR T cells in multiple myeloma and several solid tumors and highlight challenges and opportunities afforded by the current state of CAR T-cell therapies, including strategies to overcome inhibitory aspects of the tumor microenvironment and enhance antitumor efficacy., (Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
42. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date.
- Author
-
Park JH, Geyer MB, and Brentjens RJ
- Subjects
- Animals, B-Lymphocytes metabolism, B-Lymphocytes pathology, Hematologic Neoplasms genetics, Hematologic Neoplasms metabolism, Humans, Adoptive Transfer, Antigens, CD19, Hematologic Neoplasms therapy, Receptors, Antigen, T-Cell genetics, T-Lymphocytes transplantation
- Abstract
Adoptive transfer of T cells genetically modified to express chimeric antigen receptors (CARs) targeting CD19 has produced impressive results in treating patients with B-cell malignancies. Although these CAR-modified T cells target the same antigen, the designs of CARs vary as well as several key aspects of the clinical trials in which these CARs have been studied. It is unclear whether these differences have any impact on clinical outcome and treatment-related toxicities. Herein, we review clinical results reflecting the investigational use of CD19-targeted CAR T-cell therapeutics in patients with B-cell hematologic malignancies, in light of differences in CAR design and production, and outline the limitations inherent in comparing outcomes between studies., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
43. Reduced Toxicity Conditioning and Allogeneic Hematopoietic Progenitor Cell Transplantation for Recessive Dystrophic Epidermolysis Bullosa.
- Author
-
Geyer MB, Radhakrishnan K, Giller R, Umegaki N, Harel S, Kiuru M, Morel KD, LeBoeuf N, Kandel J, Bruckner A, Fabricatore S, Chen M, Woodley D, McGrath J, Baxter-Lowe L, Uitto J, Christiano AM, and Cairo MS
- Subjects
- Alemtuzumab, Antibodies, Monoclonal, Humanized therapeutic use, Busulfan therapeutic use, Child, Collagen Type VII genetics, Collagen Type VII metabolism, Epidermolysis Bullosa Dystrophica genetics, Epithelial Cells metabolism, Fibroblasts metabolism, Humans, Immunosuppressive Agents therapeutic use, Infant, Male, Mutation, Myeloablative Agonists therapeutic use, RNA, Messenger metabolism, Transplantation, Homologous, Vidarabine analogs & derivatives, Vidarabine therapeutic use, Epidermolysis Bullosa Dystrophica therapy, Hematopoietic Stem Cell Transplantation, Transplantation Conditioning
- Abstract
Recessive dystrophic epidermolysis bullosa is a severe, incurable, inherited blistering disease caused by COL7A1 mutations. Emerging evidence suggests hematopoietic progenitor cells (HPCs) can be reprogrammed into skin; HPC-derived cells can restore COL7 expression in COL7-deficient mice. We report two children with recessive dystrophic epidermolysis bullosa treated with reduced-toxicity conditioning and HLA-matched HPC transplantation., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
44. Enhanced survival during experimental Listeria monocytogenes sepsis in neonatal mice prophylactically treated with Th1 and macrophage immunoregulatory cytokines and mediators.
- Author
-
Geyer MB, Radhakrishnan K, Van de Ven C, Suri MS, Ayello J, and Cairo MS
- Subjects
- Animals, Animals, Newborn, Chemoprevention methods, Disease Models, Animal, Macrophages drug effects, Mice, Inbred C57BL, Sepsis immunology, Survival Analysis, Th1 Cells drug effects, Treatment Outcome, Cytokines therapeutic use, Immunotherapy methods, Listeria monocytogenes immunology, Macrophages immunology, Sepsis drug therapy, Sepsis prevention & control, Th1 Cells immunology
- Abstract
Background: Impairments in T-cell and macrophage-mediated host defense lead to increased infection-related morbidity and mortality in neonates, partly because of immaturity in T helper (Th)1 function. Listeria monocytogenes (Lm) is an intracellular pathogen disproportionately causing severe disease among neonates and the immunocompromised. Intact macrophage and Th1-mediated immune responses are critical for Lm clearance. We and others have previously demonstrated downregulation of Th1 and macrophage immunoregulatory cytokines in cord blood versus adult peripheral blood. We sought to determine whether therapeutic or prophylactic single agent or combination recombinant murine interleukin (rmIL)-12, rmIL-18, recombinant murine macrophage-colony stimulating factor (rmM-CSF), recombinant murine granulocyte macrophage-colony stimulating factor (rmGM-CSF) and recombinant murine interferon (rmIFN)-γ would enhance survival during experimental neonatal Lm sepsis., Methods: A 90% lethal dose (LD90) of Lm was established in C57/BL/6 neonatal mice. rmIL-12, rmIL-18, rmM-CSF, rmGM-CSF and rmIFN-γ were administered singly or sequentially, before or after LD90 Lm inoculation; ampicillin was administered 24 hours after inoculation., Results: Therapeutic doses of rmIL-12, rmIL-18, rmM-CSF, rmGM-CSF and rmIFN-γ as single agents and sequential therapy with rmM-CSF + (rmIL-12 and/or rmIL-18) + rmIFN-γ in addition to ampicillin were not associated with increased survival. However, prophylactic single doses of rmIL-12, rmIL-18, rmM-CSF and rmIFN-γ and prophylactic sequential doses of rmM-CSF + (rmIL-12 and/or rmIL-18) + rmIFN-γ in addition to ampicillin were associated with significantly enhanced survival compared with ampicillin alone., Conclusions: These data suggest prophylactic administration of macrophage and Th1 immunoregulatory cytokines can potentially overcome deficits in neonatal immunity to protect against Lm.
- Published
- 2014
- Full Text
- View/download PDF
45. A comparison of bronchoalveolar lavage versus lung biopsy in pediatric recipients after stem cell transplantation.
- Author
-
Qualter E, Satwani P, Ricci A, Jin Z, Geyer MB, Alobeid B, Radhakrishnan K, Bye M, Middlesworth W, Della-Letta P, Behr G, Muniz M, van de Ven C, Harrison L, Morris E, and Cairo MS
- Subjects
- Adolescent, Child, Child, Preschool, Cohort Studies, Female, Humans, Lung Diseases etiology, Male, Bronchoalveolar Lavage methods, Hematopoietic Stem Cell Transplantation adverse effects, Lung Diseases surgery, Lung Diseases therapy, Transplantation Conditioning adverse effects, Transplantation, Homologous adverse effects
- Abstract
Bronchoalveolar lavage (BAL) has been a useful initial diagnostic tool in the evaluation of pulmonary complications after hematopoietic stem cell transplantation (HSCT); however, the diagnostic sensitivity, prevalence, and outcome after BAL versus lung biopsy (LB) in pediatric HSCT patients remains to be determined. We reviewed 193 pediatric HSCT recipients who underwent a total of 235 HSCTs. Sixty-five patients (34%) underwent a total of 101 BALs for fever, respiratory distress, and/or pulmonary infiltrates on chest radiograph and/or computed tomography scan. The 1-year probability of undergoing BAL was 43.0% after allogeneic stem cell transplantation (alloSCT) and 8.5% after autologous stem cell transplantation (autoSCT) (P = .001). Sixteen of the 193 patients (8%) patients underwent 19 LBs. The probability of undergoing LB at 1 year after HSCT was 9.3%. No grade III or IV adverse events related to either procedure were observed. Of the 101 BALs performed, 40% (n = 40) were diagnostic, with a majority revealing a bacterial pathogen. Among the 19 LBs performed, 94% identified an etiology. In multivariate analysis, myeloablative conditioning alloSCT conferred the highest risk of requiring a BAL (hazard ratio [HR],8.5; P = .0002). The probability of 2-year overall survival was 20.2% in patients who underwent BAL, 17.5% for patients who underwent biopsy, and 67.4% for patients who had neither procedure. In multivariate analysis, only the requirement of a BAL was independently associated with an increased risk of mortality (HR, 2.96; P < .0001). In summary, in this cohort of pediatric HSCT recipients, BAL and LB were used in approximately 35% and 8% of pediatric HSCTs with diagnostic yields of approximately 40% and 94%, respectively, and were both associated with poor long-term outcomes., (Copyright © 2014 American Society for Blood and Marrow Transplantation. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
46. Pilot trial of risk-adapted cyclophosphamide intensity based conditioning and HLA matched sibling and unrelated cord blood stem cell transplantation in newly diagnosed pediatric and adolescent recipients with acquired severe aplastic anemia.
- Author
-
McGuinn C, Geyer MB, Jin Z, Garvin JH, Satwani P, Bradley MB, Bhatia M, George D, Duffy D, Morris E, van de Ven C, Schwartz J, Baxter-Lowe LA, and Cairo MS
- Subjects
- Adolescent, Allografts, Anemia, Aplastic mortality, Child, Child, Preschool, Cyclophosphamide adverse effects, Disease-Free Survival, Female, Graft Rejection diagnosis, Graft Rejection mortality, Graft Rejection prevention & control, Humans, Immunosuppressive Agents adverse effects, Male, Pilot Projects, Risk Assessment, Risk Factors, Survival Rate, Anemia, Aplastic diagnosis, Anemia, Aplastic therapy, Cord Blood Stem Cell Transplantation, Cyclophosphamide administration & dosage, Immunosuppressive Agents administration & dosage, Transplantation Conditioning, Unrelated Donors
- Abstract
Background: Cyclophosphamide-based conditioning regimens and allogeneic hematopoietic stem cell transplantation (AlloHSCT) from matched related donors (MRD) has resulted in the highest survival rates in children and adolescents with acquired severe aplastic anemia (SAA). Time to transplant has consistently been associated with decreased overall survival. Reduced toxicity conditioning and AlloHSCT has been used successfully in other pediatric non-malignant diseases., Procedure: We piloted a risk-adapted AlloHSCT approach, using fludarabine and anti-thymocyte globulin based conditioning with high (200 mg/kg) and low (60 mg/kg) dose cyclophosphamide as upfront treatment in newly diagnosed pediatric patients with acquired SAA incorporating alternative donor sources, including cord blood. Average risk for non-engraftment patients with <10 transfusions received low dose cyclophosphamide (60 mg/kg); High Risk, those with ≥10 transfusions received conditioning regimen with higher intensity cyclophosphamide (200 mg/kg)., Results: Seventeen patients were enrolled and underwent AlloHSCT including 12 males and 5 females with mean age of 8 years (range 3-16), and median follow-up time of 39 months (range 1-135). Donor sources included MRD BM (6/6 [n = 9], 5/6 [n = 2]) and unrelated CB (5/6 [n = 4], 4/6 [n = 2]). Five year OS was 67.6% (37.9-85.4). Three secondary graft failures (17.6%) occurred in the low dose cyclophosphamide arm., Conclusions: Upfront treatment with risk-adapted cyclophosphamide conditioning AlloSCT is well tolerated for the management of newly diagnosed pediatric and adolescent patients with acquired SAA. However, the increased risk of graft rejection in the lower dose arm warrants additional research regarding the optimal intensity of cyclophosphamide-based conditioning regimen to reduce toxicity without increasing graft failure., (© 2014 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
47. Reduced toxicity, myeloablative conditioning with BU, fludarabine, alemtuzumab and SCT from sibling donors in children with sickle cell disease.
- Author
-
Bhatia M, Jin Z, Baker C, Geyer MB, Radhakrishnan K, Morris E, Satwani P, George D, Garvin J, Del Toro G, Zuckerman W, Lee MT, Licursi M, Hawks R, Smilow E, Baxter-Lowe LA, Schwartz J, and Cairo MS
- Subjects
- Adolescent, Adult, Alemtuzumab, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Busulfan administration & dosage, Busulfan adverse effects, Child, Child, Preschool, Female, Humans, Male, Pilot Projects, Prospective Studies, Siblings, Tissue Donors, Transplantation Chimera, Transplantation Conditioning adverse effects, Vidarabine administration & dosage, Vidarabine adverse effects, Vidarabine analogs & derivatives, Young Adult, Anemia, Sickle Cell drug therapy, Anemia, Sickle Cell therapy, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Busulfan therapeutic use, Hematopoietic Stem Cell Transplantation methods, Transplantation Conditioning methods
- Abstract
BU and CY (BU/CY; 200 mg/kg) before HLA-matched sibling allo-SCT in children with sickle cell disease (SCD) is associated with ~85% EFS but is limited by the acute and late effects of BU/CY myeloablative conditioning. Alternatives include reduced toxicity but more immunosuppressive conditioning. We investigated in a prospective single institutional study, the safety and efficacy of a reduced-toxicity conditioning (RTC) regimen of BU 12.8-16 mg/kg, fludarabine 180 mg/m(2), alemtuzumab 54 mg/m(2) (BFA) before HLA-matched sibling donor transplantation in pediatric recipients with symptomatic SCD. Eighteen patients, median age 8.9 years (2.3-20.2), M/F 15/3, 15 sibling BM and 3 sibling cord blood (CB) were transplanted. Mean whole blood and erythroid donor chimerism was 91% and 88%, at days +100 and +365, respectively. Probability of grade II-IV acute GVHD was 17%. Two-year EFS and OS were both 100%. Neurological, pulmonary and cardiovascular function were stable or improved at 2 years. BFA RTC and HLA-matched sibling BM and CB allo-SCT in pediatric recipients result in excellent EFS, long-term donor chimerism, low incidence of GVHD and stable/improved organ function.
- Published
- 2014
- Full Text
- View/download PDF
48. Rituximab pharmacokinetics in children and adolescents with de novo intermediate and advanced mature B-cell lymphoma/leukaemia: a Children's Oncology Group report.
- Author
-
Barth MJ, Goldman S, Smith L, Perkins S, Shiramizu B, Gross TG, Harrison L, Sanger W, Geyer MB, Giulino-Roth L, and Cairo MS
- Subjects
- Adolescent, Adult, Age Factors, Antibodies, Monoclonal, Murine-Derived administration & dosage, Antineoplastic Agents administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Cyclophosphamide therapeutic use, Doxorubicin therapeutic use, Drug Administration Schedule, Half-Life, Humans, L-Lactate Dehydrogenase blood, Lymphoma, B-Cell drug therapy, Lymphoma, B-Cell pathology, Methotrexate therapeutic use, Neoplasm Staging, Pilot Projects, Prednisone therapeutic use, Rituximab, Vincristine therapeutic use, Young Adult, Antibodies, Monoclonal, Murine-Derived blood, Antineoplastic Agents blood, Lymphoma, B-Cell blood
- Abstract
The ANHL01P1 trial was undertaken to determine pharmacokinetics and safety following the addition of rituximab to French-American-British/Lymphome Malins de Burkitt (FAB/LMB96) chemotherapy in 41 children and adolescents with Stage III/IV mature B-cell lymphoma/leukaemia. Patients received rituximab (375 mg/m(2) ) days -2 and 0 of two induction cycles and day 0 of two consolidation cycles. Highest peak levels were achieved following the second dose of each induction cycle [299 ± 19 and 384 ± 25 μg/ml (Group-B); 245 ± 31 and 321 ± 32 μg/ml (Group-C)] with sustained troughs and t½ of 26-29 d. Rituximab can be safely added to FAB chemotherapy with high early rituximab peak/trough levels and a long t½., (© 2013 John Wiley & Sons Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
49. Busulfan, fludarabine, and alemtuzumab conditioning and unrelated cord blood transplantation in children with sickle cell disease.
- Author
-
Radhakrishnan K, Bhatia M, Geyer MB, Del Toro G, Jin Z, Baker C, Harrison L, Morris E, Baxter-Lowe LA, and Cairo MS
- Subjects
- Alemtuzumab, Anemia, Sickle Cell immunology, Anemia, Sickle Cell pathology, Child, Child, Preschool, Female, Graft Rejection, Graft Survival drug effects, Humans, Infant, Male, Unrelated Donors, Vidarabine therapeutic use, Anemia, Sickle Cell therapy, Antibodies, Monoclonal, Humanized therapeutic use, Busulfan therapeutic use, Cord Blood Stem Cell Transplantation, Myeloablative Agonists therapeutic use, Transplantation Conditioning, Vidarabine analogs & derivatives
- Published
- 2013
- Full Text
- View/download PDF
50. The pharmacokinetics and safety of twice daily i.v. BU during conditioning in pediatric allo-SCT recipients.
- Author
-
Le Gall JB, Milone MC, Waxman IM, Shaw LM, Harrison L, Duffy D, van de Ven C, Militano O, Geyer MB, Morris E, Bhatia M, Satwani P, George D, Garvin JH, Bradley MB, Schwartz J, Baxter-Lowe LA, and Cairo MS
- Subjects
- Adolescent, Age Factors, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating blood, Busulfan administration & dosage, Busulfan adverse effects, Busulfan blood, Child, Child, Preschool, Cohort Studies, Drug Administration Schedule, Drug Therapy, Combination adverse effects, Female, Follow-Up Studies, Graft Survival drug effects, Half-Life, Humans, Infant, Infusions, Intravenous, Male, Metabolic Clearance Rate, Myeloablative Agonists administration & dosage, Myeloablative Agonists adverse effects, Myeloablative Agonists blood, Transplantation, Homologous, Antineoplastic Agents, Alkylating pharmacokinetics, Busulfan pharmacokinetics, Myeloablative Agonists pharmacokinetics, Stem Cell Transplantation adverse effects, Transplantation Conditioning adverse effects
- Abstract
Intravenous BU divided four times daily (q6 h) has been shown to be safe and effective in pediatric allo-SCT recipients. Though less frequent dosing is desirable, pharmacokinetic (PK) data on twice daily (q12 h) i.v. BU administration in pediatric allo-SCT recipients is limited. We prospectively examined the PK results in a cohort of pediatric allo-SCT recipients receiving i.v. BU q12 h as part of conditioning before allo-SCT. BU levels were obtained after the first dose of conditioning. PK parameter analysis (n=49) yielded the following 95% confidence intervals (CI₉₅): weight-normalized volume of distribution: 0.65-0.73 L/kg; t(1/2): 122-147 min; weight-normalized clearance (CL(n)): 3.4-4.3 mL/min/kg; and area under the curve: 1835-2180 mmol × min/L. From these results, a steady state concentration was calculated with CI₉₅ between 628-746 ng/mL. Comparison between recipients ≤4 vs >4 years old revealed significant differences in t(1/2) (mean: 115 vs 146 min, P=0.008) and CL(n) (mean: 4.4 vs 3.5 mL/min/kg, P=0.038). Intravenous BU q12 h had a comparable PK to i.v. BU q6 h PK seen in the literature, and in pediatric allo-SCT recipients, is a feasible, attractive alternative to i.v. q6h dosing.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.