Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Blood Adv ; 6(5): 1444-1453, 2022 03 08.
Article in English | MEDLINE | ID: mdl-34581764

ABSTRACT

We report results of our prospective pilot trial evaluating safety/feasibility of peritransplantation ruxolitinib for myelofibrosis treatment. Primary objectives were to determine safety and maximum tolerated dose (MTD) of ruxolitinib. Ruxolitinib was administered at 2 dose levels (DLs) of 5 and 10 mg twice daily, with fludarabine/melphalan conditioning regimen and tacrolimus/sirolimus graft-versus-host disease (GVHD) prophylaxis. We enrolled 6 and 12 patients at DL1 and DL2, respectively. Median age at transplantation was 65 years (range, 25-73). Per Dynamic International Prognostic Scoring System, 4 patients were high and 14 intermediate risk. Peripheral blood stem cells were graft source from matched sibling (n = 5) or unrelated (n = 13) donor. At each DL, 1 patient developed dose-limiting toxicities (DLTs): grade 3 cardiac and gastrointestinal with grade 4 pulmonary DLTs in DL1, and grade 3 kidney injury in DL2. All patients achieved engraftment. Grade 2 to 4 and 3 to 4 acute GVHD cumulative incidence was 17% (95% confidence interval [CI], 6-47) and 11% (95% CI, 3-41), respectively. Cumulative incidence of 1-year chronic GVHD was 42% (95% CI, 24-74). With 22.6-month (range, 6.2-25.8) median follow-up in surviving patients, 1-year overall and progression-free survival were 77% (95% CI, 50-91) and 71% (95% CI, 44-87), respectively. Causes of death (n = 4) were cardiac arrest, GVHD, respiratory failure, and refractory GVHD of liver. Our results show peritransplantation ruxolitinib is safe and well tolerated at MTD of 10 mg twice daily and associated with dose-dependent pharmacokinetic and cytokine profile. Early efficacy data are highly promising in high-risk older patients with myelofibrosis. This trial was registered at www.clinicaltrials.gov as #NCT02917096.


Subject(s)
Graft vs Host Disease , Primary Myelofibrosis , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Humans , Nitriles , Prospective Studies , Pyrazoles , Pyrimidines/therapeutic use
3.
Haematologica ; 106(9): 2397-2404, 2021 09 01.
Article in English | MEDLINE | ID: mdl-32732354

ABSTRACT

In a phase-2 study, the telomerase inhibitor imetelstat induced rapid hematologic responses in all patients with essential thrombocythemia who were refractory or intolerant to prior therapies. Significant molecular responses were achieved within 3-6 months in 81% of patients with phenotypic driver mutations in JAK2, CALR and MPL. Here, we investigated the dynamics of additional somatic mutations in response to imetelstat. At study entry, 50% of patients carried 1-5 additional mutations in the genes ASXL1, CBL, DNMT3A, EZH2, IDH1, SF3B1, TET2, TP53 and U2AF1. Three patients with baseline mutations also had late-emerging mutations in TP53, IDH1 and TET2. Most clones with additional mutations were responsive to imetelstat and decreased with the driver mutation, including the poor prognostic ASXL1, EZH2 and U2AF1 mutations while SF3B1 and TP53 mutations were associated with poorer molecular response. Overall, phenotypic driver mutation response was significantly deeper in patients without additional mutations (P = 0.04) and correlated with longer duration of response. In conclusion, this detailed molecular analysis of highly pretreated and partly resistant patients with essential thrombocythemia reveals a high individual patient complexity. Moreover, imetelstat demonstrates potential to inhibit efficiently co-incident mutations occurring in neoplastic clones in patients with essential thrombocythemia. (ClinicalTrials.gov number, NCT01243073. N Engl J Med 2015; 373:920-928, DOI: 10.1056/NEJMoa1503479.).


Subject(s)
Thrombocythemia, Essential , Clone Cells , Humans , Janus Kinase 2/genetics , Mutation , Oligonucleotides , Thrombocythemia, Essential/drug therapy , Thrombocythemia, Essential/genetics
4.
J Natl Compr Canc Netw ; 18(10): 1385-1415, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33022644

ABSTRACT

Chronic myeloid leukemia (CML) is defined by the presence of Philadelphia chromosome (Ph) which results from a reciprocal translocation between chromosomes 9 and 22 [t(9;22] that gives rise to a BCR-ABL1 fusion gene. CML occurs in 3 different phases (chronic, accelerated, and blast phase) and is usually diagnosed in the chronic phase. Tyrosine kinase inhibitor therapy is a highly effective first-line treatment option for all patients with newly diagnosed chronic phase CML. This manuscript discusses the recommendations outlined in the NCCN Guidelines for the diagnosis and management of patients with chronic phase CML.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukemia, Myeloid, Chronic-Phase , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Medical Oncology , Philadelphia Chromosome , Translocation, Genetic
5.
J Natl Compr Canc Netw ; 18(9): 1248-1269, 2020 09.
Article in English | MEDLINE | ID: mdl-32886902

ABSTRACT

Eosinophilic disorders and related syndromes represent a heterogeneous group of neoplastic and nonneoplastic conditions, characterized by more eosinophils in the peripheral blood, and may involve eosinophil-induced organ damage. In the WHO classification of myeloid and lymphoid neoplasms, eosinophilic disorders characterized by dysregulated tyrosine kinase (TK) fusion genes are recognized as a new category termed, myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB or FGFR1 or with PCM1-JAK2. In addition to these aforementioned TK fusion genes, rearrangements involving FLT3 and ABL1 genes have also been described. These new NCCN Guidelines include recommendations for the diagnosis, staging, and treatment of any one of the myeloid/lymphoid neoplasms with eosinophilia (MLN-Eo) and a TK fusion gene included in the 2017 WHO Classification, as well as MLN-Eo and a FLT3 or ABL1 rearrangement.


Subject(s)
Eosinophilia , Myeloproliferative Disorders , Neoplasms , Eosinophilia/diagnosis , Eosinophilia/genetics , Humans , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/therapy , Oncogene Proteins, Fusion/genetics
6.
Biol Blood Marrow Transplant ; 26(8): 1386-1393, 2020 08.
Article in English | MEDLINE | ID: mdl-32439475

ABSTRACT

As the world of cellular therapy expands to include immune effector cell (IEC) products such as commercial chimeric antigen receptor (CAR) T cells, quality management (QM) professionals are faced with creating either new IEC stand-alone programs or expand existing hematopoietic cell transplantation (HCT) programs to promote patient safety and be aligned with quality, regulatory, and accreditation requirements. The team professionals at City of Hope (COH) recently expanded the quality HCT program to include IEC products and, in doing so, implemented new regulatory infrastructure while maintaining high quality patient care. At COH, we developed the quality structure of our cellular therapy program through collaborations between quality, regulatory, and CAR T patient care committees, which included physicians and nurse coordinators. To ensure the quality of our program, we monitor data collection and reporting, perform quarterly proactive audits of, for example, outcome analysis, and measure selected end-points for benchmarking purposes. QM professionals play a critical role in the monitoring and evaluation processes and provide guidance on how to implement accreditation requirements and what impact the requirements may have on care management. Here we describe the process by which COH expanded our HCT QM program to include IEC therapy. We share examples of how we developed our overall program structure and other key items such as how we addressed patient care management and accreditation to apprise other programs that wish to create and/or expand existing programs.


Subject(s)
Hematopoietic Stem Cell Transplantation , Receptors, Chimeric Antigen , Accreditation , Humans , Quality of Health Care , T-Lymphocytes
7.
Cancer ; 125(22): 4033-4042, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31412155

ABSTRACT

BACKGROUND: Late mortality was investigated in patients with chronic myelogenous leukemia (CML) who underwent blood or bone marrow transplant (BMT) with or without prior tyrosine kinase inhibitor (TKI) therapy. METHODS: By using data from the Blood or Marrow Transplant Survivor Study, the authors examined late mortality in 447 patients with CML who underwent BMT between 1974 and 2010, conditional on surviving ≥2 years post-BMT. For vital status information, the medical records, the National Death Index, and the Accurint database were used. Standardized mortality ratios (SMRs) were calculated using general population age-specific, sex-specific, and calendar-specific mortality rates. Kaplan-Meier techniques and Cox regression were used for all-cause mortality analyses. Cumulative incidence and proportional subdistribution hazards models for competing risks were used for cause-specific mortality analyses. RESULTS: The 10-year overall survival rate was 65.7% and 73% for those who underwent transplant with and without pre-BMT exposure to TKI therapy, respectively. Patients who underwent transplant with and without pre-BMT TKI experienced SMRs of 6.4 and 6.4, respectively (P = .8); and the SMRs were 11.6 and 8.1, respectively, for those with high-risk disease (P = .2). Independent predictors of non-CML-related mortality included chronic graft-versus-host disease (hazard ratio [HR], 2.8; 95% CI, 1.8-4.4) and busulfan/cyclophosphamide conditioning (HR, 0.5; 95% CI, 0.3-0.9; reference, total body irradiation/cyclophosphamide conditioning). The 20-year cumulative incidence of CML-related and non-CML-related mortality was 6% and 36%, respectively, for the entire cohort. Both CML-related mortality (HR, 1.0; 95% CI, 0.1-12.6) and non-CML-related mortality (HR, 1.3; 95% CI, 0.6-3.1) were comparable for those with and without pre-BMT TKI therapy. CONCLUSIONS: The similar late mortality experienced by patients with CML who undergo transplantation with or without pre-BMT TKIs suggests that allogeneic BMT can be considered in the context of TKI intolerance or nonadherence. The prevention of post-BMT non-CML-related mortality could favorably affect long-term survival.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Adolescent , Adult , Aged , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/methods , Cause of Death , Child , Child, Preschool , Combined Modality Therapy , Female , Graft vs Host Disease/etiology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Male , Middle Aged , Prognosis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Transplantation, Homologous , Treatment Outcome , Young Adult
8.
Case Rep Infect Dis ; 2019: 5464230, 2019.
Article in English | MEDLINE | ID: mdl-31032127

ABSTRACT

A 69-year-old immunocompromised man developed mitral valve endocarditis due to Salmonella enterica serotype Mbandaka, contracted from the cereal outbreak. The patient had a history of HLA-matched related hematopoietic stem cell transplant with persistent graft-versus-host disease (GVHD). This case report discusses prior international outbreaks that occurred due to Salmonella enterica subtype Mbandaka, the risks of developing endovascular infections from salmonellosis, and persistent infections that may develop more frequently with S. enterica serotype Mbandaka. The patient received a six-week course of intravenous antibiotics and remains on oral suppressive antibiotics, with his length of therapy to be determined based on his GVHD treatment.

9.
Haematologica ; 104(1): 93-101, 2019 01.
Article in English | MEDLINE | ID: mdl-30093398

ABSTRACT

Dasatinib, a second-generation BCR-ABL1 tyrosine kinase inhibitor, is approved for the treatment of chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia, both as first-line therapy and after imatinib intolerance or resistance. While generally well tolerated, dasatinib has been associated with a higher risk for pleural effusions. Frequency, risk factors, and outcomes associated with pleural effusion were assessed in two phase 3 trials (DASISION and 034/Dose-optimization) and a pooled population of 11 trials that evaluated patients with chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia treated with dasatinib (including DASISION and 034/Dose-optimization). In this largest assessment of patients across the dasatinib clinical trial program (N=2712), pleural effusion developed in 6-9% of patients at risk annually in DASISION, and in 5-15% of patients at risk annually in 034/Dose-optimization. With a minimum follow up of 5 and 7 years, drug-related pleural effusion occurred in 28% of patients in DASISION and in 33% of patients in 034/Dose-optimization, respectively. A significant risk factor identified for developing pleural effusion by a multivariate analysis was age. We found that overall responses to dasatinib, progression-free survival, and overall survival were similar in patients who developed pleural effusion and in patients who did not. clinicaltrials.gov identifier 00481247; 00123474.


Subject(s)
Dasatinib/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Pleural Effusion, Malignant , Adult , Disease-Free Survival , Female , Humans , Incidence , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Middle Aged , Pleural Effusion, Malignant/drug therapy , Pleural Effusion, Malignant/mortality , Pleural Effusion, Malignant/pathology , Risk Factors , Survival Rate
10.
J Natl Compr Canc Netw ; 16(12): 1500-1537, 2018 12.
Article in English | MEDLINE | ID: mdl-30545997

ABSTRACT

Mastocytosis is a group of heterogeneous disorders resulting from the clonal proliferation of abnormal mast cells and their accumulation in the skin and/or in various extracutaneous organs. Systemic mastocytosis is the most common form of mastocytosis diagnosed in adults, characterized by mast cell infiltration of one or more extracutaneous organs (with or without skin involvement). The identification of KIT D816V mutation and the emergence of novel targeted therapies have significantly improved the diagnosis and treatment of systemic mastocytosis. However, certain aspects of clinical care, particularly the diagnosis, assessment, and management of mediator-related symptoms continue to present challenges. This manuscript discusses the recommendations outlined in the NCCN Guidelines for the diagnosis and management of patients with systemic mastocytosis.


Subject(s)
Anaphylaxis/therapy , Mastocytosis, Systemic/therapy , Medical Oncology/standards , Patient Care Team/standards , Anaphylaxis/diagnosis , Anaphylaxis/immunology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Biopsy , Bone Marrow/drug effects , Bone Marrow/pathology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/standards , Histamine Antagonists/pharmacology , Histamine Antagonists/therapeutic use , Humans , Immunophenotyping/methods , Immunophenotyping/standards , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Mastocytosis, Systemic/diagnosis , Mastocytosis, Systemic/genetics , Mastocytosis, Systemic/immunology , Medical Oncology/methods , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/standards , Mutation , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins c-kit/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Societies, Medical/standards , Transplantation, Homologous/methods , Transplantation, Homologous/standards , Treatment Outcome , mRNA Cleavage and Polyadenylation Factors/genetics
11.
J Natl Compr Canc Netw ; 16(9): 1108-1135, 2018 09.
Article in English | MEDLINE | ID: mdl-30181422

ABSTRACT

Chronic myeloid leukemia (CML) is defined by the presence of Philadelphia chromosome (Ph), resulting from a reciprocal translocation between chromosomes 9 and 22 [t(9;22] that gives rise to a BCR-ABL1 fusion gene. CML occurs in 3 different phases (chronic, accelerated, and blast phase) and is usually diagnosed in the chronic phase. Tyrosine kinase inhibitor (TKI) therapy is a highly effective first-line treatment option for all patients with newly diagnosed chronic phase CML (CP-CML). The selection TKI therapy should be based on the risk score, toxicity profile of TKI, patient's age, ability to tolerate therapy, and the presence of comorbid conditions. This manuscript discusses the recommendations outlined in the NCCN Guidelines for the diagnosis and management of patients with CP-CML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Medical Oncology/standards , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/standards , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/isolation & purification , Bone Marrow/pathology , Clinical Trials as Topic , Disease Progression , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/isolation & purification , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Medical Oncology/methods , Patient Selection , Philadelphia Chromosome , Prognosis , Progression-Free Survival , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/standards , Real-Time Polymerase Chain Reaction/standards , Risk Assessment/methods , Risk Assessment/standards , Societies, Medical/standards , United States
12.
Nat Med ; 24(4): 450-462, 2018 05.
Article in English | MEDLINE | ID: mdl-29505034

ABSTRACT

Leukemia stem cells (LSCs) in individuals with chronic myelogenous leukemia (CML) (hereafter referred to as CML LSCs) are responsible for initiating and maintaining clonal hematopoiesis. These cells persist in the bone marrow (BM) despite effective inhibition of BCR-ABL kinase activity by tyrosine kinase inhibitors (TKIs). Here we show that although the microRNA (miRNA) miR-126 supported the quiescence, self-renewal and engraftment capacity of CML LSCs, miR-126 levels were lower in CML LSCs than in long-term hematopoietic stem cells (LT-HSCs) from healthy individuals. Downregulation of miR-126 levels in CML LSCs was due to phosphorylation of Sprouty-related EVH1-domain-containing 1 (SPRED1) by BCR-ABL, which led to inhibition of the RAN-exportin-5-RCC1 complex that mediates miRNA maturation. Endothelial cells (ECs) in the BM supply miR-126 to CML LSCs to support quiescence and leukemia growth, as shown using mouse models of CML in which Mir126a (encoding miR-126) was conditionally knocked out in ECs and/or LSCs. Inhibition of BCR-ABL by TKI treatment caused an undesired increase in endogenous miR-126 levels, which enhanced LSC quiescence and persistence. Mir126a knockout in LSCs and/or ECs, or treatment with a miR-126 inhibitor that targets miR-126 expression in both LSCs and ECs, enhanced the in vivo anti-leukemic effects of TKI treatment and strongly diminished LSC leukemia-initiating capacity, providing a new strategy for the elimination of LSCs in individuals with CML.


Subject(s)
Bone Marrow/pathology , Cell Self Renewal , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , Stem Cell Niche , Animals , Down-Regulation/genetics , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Gene Silencing , Hematopoietic Stem Cells/metabolism , Humans , Mice , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/ultrastructure , Protein Kinase Inhibitors/pharmacology
13.
J Natl Compr Canc Netw ; 15(10): 1193-1207, 2017 10.
Article in English | MEDLINE | ID: mdl-28982745

ABSTRACT

Myeloproliferative neoplasms (MPNs) are a group of heterogeneous disorders of the hematopoietic system that include myelofibrosis (MF), polycythemia vera (PV), and essential thrombocythemia (ET). PV and ET are characterized by significant thrombohemorrhagic complications and a high risk of transformation to MF and acute myeloid leukemia. The diagnosis and management of PV and ET has evolved since the identification of mutations implicated in their pathogenesis. These NCCN Guideline Insights discuss the recommendations outlined in the NCCN Guidelines for the risk stratification, treatment, and special considerations for the management of PV and ET.


Subject(s)
Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/therapy , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Humans , Risk Assessment , Treatment Outcome
14.
Haematologica ; 102(3): 519-528, 2017 03.
Article in English | MEDLINE | ID: mdl-27927766

ABSTRACT

A vailable tyrosine kinase inhibitors for chronic myeloid leukemia bind in an adenosine 5'-triphosphate-binding pocket and are affected by evolving mutations that confer resistance. Rebastinib was identified as a switch control inhibitor of BCR-ABL1 and FLT3 and may be active against resistant mutations. A Phase 1, first-in-human, single-agent study investigated rebastinib in relapsed or refractory chronic or acute myeloid leukemia. The primary objectives were to investigate the safety of rebastinib and establish the maximum tolerated dose and recommended Phase 2 dose. Fifty-seven patients received treatment with rebastinib. Sixteen patients were treated using powder-in-capsule preparations at doses from 57 mg to 1200 mg daily, and 41 received tablet preparations at doses of 100 mg to 400 mg daily. Dose-limiting toxicities were dysarthria, muscle weakness, and peripheral neuropathy. The maximum tolerated dose was 150 mg tablets administered twice daily. Rebastinib was rapidly absorbed. Bioavailability was 3- to 4-fold greater with formulated tablets compared to unformulated capsules. Eight complete hematologic responses were achieved in 40 evaluable chronic myeloid leukemia patients, 4 of which had a T315I mutation. None of the 5 patients with acute myeloid leukemia responded. Pharmacodynamic analysis showed inhibition of phosphorylation of substrates of BCR-ABL1 or FLT3 by rebastinib. Although clinical activity was observed, clinical benefit was insufficient to justify continued development in chronic or acute myeloid leukemia. Pharmacodynamic analyses suggest that other kinases inhibited by rebastinib, such as TIE2, may be more relevant targets for the clinical development of rebastinib (clinicaltrials.gov Identifier:00827138).


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/administration & dosage , Quinolines/administration & dosage , Adult , Aged , Aged, 80 and over , Drug Monitoring , Drug Resistance, Neoplasm/genetics , Female , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myeloid, Acute/genetics , Male , Maximum Tolerated Dose , Middle Aged , Mutation , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Quinolines/adverse effects , Quinolines/pharmacokinetics , Treatment Outcome , Young Adult
15.
J Natl Compr Canc Netw ; 14(12): 1505-1512, 2016 12.
Article in English | MEDLINE | ID: mdl-27956535

ABSTRACT

The NCCN Guidelines for Chronic Myeloid Leukemia (CML) provide recommendations for the management of chronic-phase and advanced-phase CML in adult patients. The median age of disease onset is 67 years. However, because CML occurs in all age groups, clinical care teams should be prepared to address issues relating to fertility and pregnancy with patients who are of reproductive age at the time of diagnosis. CML is relatively rare in children and there are no evidence-based recommendations for the management of CML in pediatric population. These NCCN Guidelines Insights discuss special considerations for the management of CML during pregnancy and for the management of CML in the pediatric population.


Subject(s)
Abnormalities, Drug-Induced/epidemiology , Fertility/drug effects , Leukemia, Myeloid, Chronic-Phase/drug therapy , Pregnancy Complications, Neoplastic/drug therapy , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Child , Evidence-Based Medicine/standards , Female , Humans , Practice Guidelines as Topic , Pregnancy , Prognosis , Protein Kinase Inhibitors/administration & dosage , Withholding Treatment
16.
J Natl Compr Canc Netw ; 14(12): 1572-1611, 2016 12.
Article in English | MEDLINE | ID: mdl-27956542

ABSTRACT

Myelofibrosis (MF), polycythemia vera (PV), and essential thrombocythemia (ET) are a group of heterogeneous disorders of the hematopoietic system collectively known as Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs). The diagnosis and the management of patients with MPNs have evolved since the identification of mutations that activate the JAK pathway (JAK2, CALR, and MPL mutations) and the development of targeted therapies has resulted in significant improvements in disease-related symptoms and quality of life. This manuscript discusses the recommendations outlined in the NCCN Guidelines for the diagnostic workup of MPN (MF, PV, and ET), risk stratification, treatment, and supportive care strategies for the management of MF.


Subject(s)
Medical Oncology/standards , Polycythemia Vera , Primary Myelofibrosis , Thrombocythemia, Essential , Calreticulin/genetics , Calreticulin/metabolism , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mutation , Philadelphia Chromosome , Polycythemia Vera/diagnosis , Polycythemia Vera/epidemiology , Polycythemia Vera/genetics , Polycythemia Vera/therapy , Prevalence , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/epidemiology , Primary Myelofibrosis/genetics , Primary Myelofibrosis/therapy , Quality of Life , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Risk Assessment , Signal Transduction , Thrombocythemia, Essential/diagnosis , Thrombocythemia, Essential/epidemiology , Thrombocythemia, Essential/genetics , Thrombocythemia, Essential/therapy
17.
Clin Lymphoma Myeloma Leuk ; 16(9): 519-522, 2016 09.
Article in English | MEDLINE | ID: mdl-27394652

ABSTRACT

BACKGROUND: The outcome of patients with acute lymphoblastic leukemia (ALL) relapsing after allogeneic hematopoietic cell transplantation (AlloHCT) is poor. Although morphologic remission can sometimes be achieved, such remissions are usually transient if not consolidated by a second AlloHCT (AlloHCT2). MATERIALS AND METHODS: We retrospectively analyzed the outcomes of 27 patients with ALL who had undergone AlloHCT2 for relapsed disease at our center during a 12-year period. RESULTS: With a median follow-up of 50.9 months for living patients, the 2-year overall and event-free survival were 40.7% and 29.6%, respectively. Patients with either a disease-free interval or interval between transplants of > 1 year had better overall survival (P = .02 and P = .0005) after AlloHCT2. CONCLUSION: AlloHCT2 remains a potential curative option in a subset of patients with relapsed ALL after the first AlloHCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Child , Female , Follow-Up Studies , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Recurrence , Retreatment , Retrospective Studies , Survival Analysis , Tissue Donors , Transplantation Conditioning , Transplantation, Homologous , Treatment Outcome , Young Adult
18.
Lancet Haematol ; 3(2): e87-98, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26853648

ABSTRACT

BACKGROUND: Patients seropositive for cytomegalovirus (CMV) and undergoing allogeneic haemopoietic stem-cell transplantation (HCT) are at risk for CMV reactivation. Stimulating viral immunity by vaccination might achieve CMV viraemia control without the need for antiviral agents. CMVPepVax is a chimeric peptide composed of a cytotoxic CD8 T-cell epitope from CMV pp65 and a tetanus T-helper epitope. It is formulated with the adjuvant PF03512676, a Toll-like receptor 9 agonist, which augments cellular immunity. We aimed to assess safety, immunogenicity, and possible clinical benefit of the CMVPepVax vaccine in patients undergoing HCT. METHODS: We did a randomised, open-label, phase 1b trial at one transplant centre in the USA. Eligible patients were CMV-seropositive, positive for HLA-A*0201, aged 18-75 years, and undergoing HCT from a matched-related or matched-unrelated donor. Patients were reassessed for eligibility on day 28 after HCT. We randomly allocated patients to either the CMVPepVax vaccine or observation, in blocks stratified by CMV donor serostatus. CMVPepVax was administered subcutaneously on days 28 and 56. The primary outcome was safety, which consisted of secondary graft failure, grade III-IV acute GVHD, non-relapse mortality by day 100, serious adverse events related to the vaccine (judged by the data and safety monitoring committee [DSMC]) grade 3-4 adverse events related to the vaccine (judged by the DSMC) within 2 weeks of vaccination, and development of double-strand (ds) DNA autoantibodies. Statistical analyses included all randomised patients and were done per-protocol. This study is registered with ClinicalTrials.gov, number NCT01588015. This trial is closed to accrual and the final analysis is presented in this report. FINDINGS: Between Oct 31, 2012, and Nov 5, 2014, 36 eligible patients were allocated to either CMVPepVax (n=18) or observation (n=18), with no adverse effect on HCT (no secondary graft failures in either group) or cases of acute GVHD (seven patients assigned vaccine and six under observation had acute GVHD of grade 2 or less), and no unexpected adverse events. Compared with observation, better relapse-free survival was recorded in patients allocated the vaccine (seven vs one; hazard ratio [HR] 0·12, 95% CI 0·01-0·94; p=0·015). No patients had non-relapse mortality by day 100. One serious adverse event (grade 1 fever) was attributed to CMVPepVax but resolved within 48 h. Four patients assigned the vaccine had a serious adverse event, which was unrelated to the vaccine (grade 3 thrombocytopenia, grade 3 device-related infection, grade 2 nausea, and grade 1 fever), compared with nine patients under observation (grade 4 maculopapular rash, grade 3 nausea, grade 3 infection, grade 3 thrombotic thrombocytopenic purpurea, grade 2 nausea, grade 2 generalised muscle weakness, grade 2 infection, grade 1 fever, and grade 1 fatigue; p=0·16). 54 grade 3-4 adverse events were reported in patients assigned the vaccine compared with 91 in patients who were under observation (p=0·2). No patients had grade III-IV acute GVHD or developed dsDNA autoantibodies. INTERPRETATION: The results show safety and immunogenicity of the CMVPepVax vaccine. The prospect of substantial clinical benefits warrant testing in a phase 2 trial. FUNDING: National Cancer Institute.


Subject(s)
Cytomegalovirus Infections/prevention & control , Hematopoietic Stem Cell Transplantation , Viral Vaccines/therapeutic use , Viremia/prevention & control , Adjuvants, Immunologic/administration & dosage , Adult , Aged , Cytomegalovirus , Disease-Free Survival , Epitopes, T-Lymphocyte/immunology , Female , Graft vs Host Disease , Humans , Male , Middle Aged , Oligodeoxyribonucleotides/administration & dosage , Toll-Like Receptor 9/agonists , Treatment Outcome , Vaccines, Synthetic/therapeutic use , Virus Activation , Young Adult
20.
N Engl J Med ; 373(10): 920-8, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26332546

ABSTRACT

BACKGROUND: Imetelstat, a 13-mer oligonucleotide that is covalently modified with lipid extensions, competitively inhibits telomerase enzymatic activity. It has been shown to inhibit megakaryocytic proliferation in vitro in cells obtained from patients with essential thrombocythemia. In this phase 2 study, we investigated whether imetelstat could elicit hematologic and molecular responses in patients with essential thrombocythemia who had not had a response to or who had had unacceptable side effects from prior therapies. METHODS: A total of 18 patients in two sequential cohorts received an initial dose of 7.5 or 9.4 mg of imetelstat per kilogram of body weight intravenously once a week until attainment of a platelet count of approximately 250,000 to 300,000 per cubic millimeter. The primary end point was the best hematologic response. RESULTS: Imetelstat induced hematologic responses in all 18 patients, and 16 patients (89%) had a complete hematologic response. At the time of the primary analysis, 10 patients were still receiving treatment, with a median follow-up of 17 months (range, 7 to 32 [ongoing]). Molecular responses were seen in 7 of 8 patients who were positive for the JAK2 V617F mutation (88%; 95% confidence interval, 47 to 100). CALR and MPL mutant allele burdens were also reduced by 15 to 66%. The most common adverse events during treatment were mild to moderate in severity; neutropenia of grade 3 or higher occurred in 4 of the 18 patients (22%) and anemia, headache, and syncope of grade 3 or higher each occurred in 2 patients (11%). All the patients had at least one abnormal liver-function value; all persistent elevations were grade 1 or 2 in severity. CONCLUSIONS: Rapid and durable hematologic and molecular responses were observed in patients with essential thrombocythemia who received imetelstat. (Funded by Geron; ClinicalTrials.gov number, NCT01243073.).


Subject(s)
Indoles/administration & dosage , Niacinamide/analogs & derivatives , Telomerase/antagonists & inhibitors , Thrombocythemia, Essential/drug therapy , Aged , Aged, 80 and over , Drug Administration Schedule , Female , Humans , Indoles/adverse effects , Infusions, Intravenous , Janus Kinase 2/genetics , Liver/enzymology , Liver Function Tests , Male , Middle Aged , Mutation , Niacinamide/administration & dosage , Niacinamide/adverse effects , Oligonucleotides , Pilot Projects , Thrombocythemia, Essential/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...