Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Am J Med Genet A ; 176(12): 2924-2929, 2018 12.
Article in English | MEDLINE | ID: mdl-30302932

ABSTRACT

This report summarizes and highlights the fifth International RASopathies Symposium: When Development and Cancer Intersect, held in Orlando, Florida in July 2017. The RASopathies comprise a recognizable pattern of malformation syndromes that are caused by germ line mutations in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) pathway. Because of their common underlying pathogenetic etiology, there is significant overlap in their phenotypic features, which includes craniofacial dysmorphology, cardiac, cutaneous, musculoskeletal, gastrointestinal and ocular abnormalities, neurological and neurocognitive issues, and a predisposition to cancer. The RAS pathway is a well-known oncogenic pathway that is commonly found to be activated in somatic malignancies. As in somatic cancers, the RASopathies can be caused by various pathogenetic mechanisms that ultimately impact or alter the normal function and regulation of the MAPK pathway. As such, the RASopathies represent an excellent model of study to explore the intersection of the effects of dysregulation and its consequence in both development and oncogenesis.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , ras Proteins/genetics , Animals , Gene Expression Regulation , Genetic Association Studies/methods , Human Development , Humans , Models, Biological , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Organogenesis/genetics , Signal Transduction , Syndrome , ras Proteins/metabolism
2.
Leuk Res ; 94: 106369, 2020 07.
Article in English | MEDLINE | ID: mdl-32442785

ABSTRACT

Phase 1 results from a Phase 1/2 study comprise 18 patients with myelodysplastic syndromes (MDS; n = 9), acute myeloid leukemia (AML; n = 8), and chronic myelomonocytic leukemia (CMML; n = 1) who were either hypomethylating agent naïve (n = 10) or relapsed/refractory following prior hypomethylating agent therapy (n = 8) (NCT01926587). Patients received oral rigosertib, an inhibitor of Ras-effector pathways, in 3 successive cohorts (140 mg twice daily, 280 mg twice daily, or 840 mg/day [560 mg morning/280 mg evening]) for 3 weeks of a 4-week cycle. Patients received parenteral azacitidine (75 mg/m2/day × 7 days) during the second week; the cycle repeated every 4 weeks. The combination was well tolerated for a median of 4 (range 1-41) cycles, with 72% of patients experiencing ≥1 serious adverse events. No dose-limiting toxicities were observed. Thus, no maximum tolerated dose was reached. The most frequently reported adverse events were diarrhea (50%), constipation, fatigue, and nausea (each 44%), and pneumonia and back pain (each 33%). Sequential administration demonstrated an overall response rate of 56% in evaluable patients, with responses observed in 7/9 MDS/CMML patients (78%) and 2/7 AML patients (29%). Further clinical studies are warranted to investigate this doublet therapy in patients with myeloid malignancies.


Subject(s)
Azacitidine/administration & dosage , Glycine/analogs & derivatives , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Sulfones/administration & dosage , Adult , Aged , Aged, 80 and over , Azacitidine/adverse effects , Female , Glycine/administration & dosage , Glycine/adverse effects , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/pathology , Sulfones/adverse effects
3.
Leuk Res ; 64: 10-16, 2018 01.
Article in English | MEDLINE | ID: mdl-29144985

ABSTRACT

This Phase 1/2, dose-escalating study of rigosertib enrolled 22 patients with higher-risk myelodysplastic syndromes (MDS) (n=9) and acute myeloid leukemia (AML; n=13) who had relapsed or were refractory to standard therapy and for whom no second-line therapies were approved. Patients received 3- to 7-day continuous intravenous infusions of rigosertib, an inhibitor of Ras-effector pathways that interacts with the Ras-binding domains, common to several signaling proteins including Raf and PI3 kinase. Rigosertib was administered at doses of 650-1700mg/m2/day in 14-day cycles. Initial dose escalation followed a Fibonacci scheme, followed by recommended phase 2 dose confirmation in an expanded cohort. Rigosertib was well tolerated for up to 23 cycles, with no treatment-related deaths and 18% of patients with related serious adverse events (AEs). Common AEs were fatigue, diarrhea, pyrexia, dyspnea, insomnia, and anemia. Rigosertib exhibited biologic activity, with reduction or stabilization of bone marrow blasts and improved peripheral blood counts in a subset of patients. Ten of 19 evaluable patients (53%) demonstrated bone marrow/peripheral blood responses (n=4 MDS, n=1 AML) or stable disease (n=3 MDS, n=2 AML). Median survival was 15.7 and 2.0 months for responders and non-responders, respectively. Additional studies of rigosertib are ongoing in higher-risk MDS (NCT00854646).


Subject(s)
Antineoplastic Agents/administration & dosage , Glycine/analogs & derivatives , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Sulfones/administration & dosage , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Disease Progression , Dose-Response Relationship, Drug , Female , Glycine/administration & dosage , Glycine/adverse effects , Humans , Infusions, Intravenous , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/mortality , Male , Maximum Tolerated Dose , Middle Aged , Sulfones/adverse effects , Survival Analysis
4.
Leuk Res ; 29(5): 481-91, 2005 May.
Article in English | MEDLINE | ID: mdl-15755500

ABSTRACT

Appropriate treatment for nonreactive thrombocytosis resulting from a myeloproliferative disorder (MPD) is surrounded by controversy. Although few doubt the association of thrombocytosis with increased risk for life-threatening events such as thrombosis or hemorrhage, or the association between clonal myeloproliferation and the progression to acute leukemia or myelofibrosis, controversy exists regarding the timing and nature of appropriate therapeutic intervention. Studies have shown that treatment with myelosuppressive agents such as chlorambucil, busulfan, radiophosphorus (32P), and hydroxyurea reduces the platelet count. However, investigators have also identified an increased risk of drug-related leukemic transformation. An ideal cytoreductive treatment for long-term use should minimize thrombosis and avoid long-term complications, especially acute leukemia (AL). Anagrelide, an imidazoquinolin, inhibits megakaryopoiesis and more selectively reduces platelet production in humans. A retrospective analysis of an open-label, multicenter, international trial reviewing 3660 anagrelide-treated patients was performed to assess efficacy and long-term safety, specifically potential for increased leukemogenicity. The study included MPD patients with thrombocytosis diagnosed according to Polycythemia Vera Study Group (PVSG) criteria. Of all patients enrolled, 81% had previously received other myelosuppressive agents; of these, 33% were transferred from the original agent to anagrelide due to toxicity and 31% were transferred because of poor platelet control. Over 45% of patients were symptomatic due to thrombocythemia, most commonly vascular sequelae (25%). Dosage was titrated to achieve a platelet count < 600 x 10(9) L(-1) and ideally between 130 and 450 x 10(9) L(-1). The safety cohort of 3660 patients, including 2251 with essential thrombocythemia (ET), 462 with polycythemia vera (PV), and 947 with chronic myeloid leukemia (CML) and other MPDs, was analyzed to establish the incidence of leukemic transformation in patients with ET and PV. From the Safety Population, 12.8% (467/3660) of patients were treated with anagrelide as the sole cytoreductive agent (naive patients). Acute leukemia/myelodysplasia developed in 2.1% of ET patients (47/2251) with a maximum follow-up of 7.1 years. Of the PV patients, 2.8% developed acute leukemia/myelodysplastic syndrome (13/462), with a maximum follow-up of 7.0 years. ET and PV patients who transformed to AL had all been previously exposed to other cytotoxics; there were no ET or PV patients in the study who transformed to AL exposed solely to anagrelide. With maximum follow-up over 7 years, anagrelide achieved platelet control in over 75% of MPD patients and did not increase the conversion to acute leukemia during the treatment duration analyzed. Longer follow-up is required to confirm these important observations regarding the long-term safety of anagrelide.


Subject(s)
Myeloproliferative Disorders/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Quinazolines/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Blood Platelets/drug effects , Blood Platelets/metabolism , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Myeloproliferative Disorders/pathology , Retrospective Studies , Safety , Time Factors
5.
Semin Hematol ; 40(1 Suppl 1): 30-3, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12682880

ABSTRACT

Myelofibrosis with myeloid metaplasia, also known as idiopathic myelofibrosis (IF) or agnogenic myeloid metaplasia, is one of the characteristic manifestations of polycythemia vera (PV) in the spent phase, and has a particularly adverse prognosis. IF may also present de novo. To date, treatment strategies for both spent-phase PV and IF have frustrated both clinicians and patients, with little clear progress made over the past 50 years. Treatment modalities with some benefit in chronic myeloid leukemia (CML), such as interferon (IFN), have been used to shrink the massive organomegaly seen in these patients and to improve their marrow function, but are not curative, and not all patients respond or can tolerate the agent. A curative approach is allogeneic peripheral hematopoietic stem cell transplantation. The preparative regimens used in fully ablative techniques rule out older patients for consideration, and many younger patients with good prognostic criteria may do sufficiently well on medical treatment or observation to avoid transplantation. Older patients may have the option to undergo a human leukocyte antigen (HLA)-identical sibling transplant using a reduced intensity preparative regimen in order to minimize peritransplant mortality. Thus a prerequisite to the broad use of transplantation is objective determination of candidacy. Several evaluation methods agree that anemia, age, and cytogenetic abnormalities all predict poor survival in IF, suggesting that patients with anemia and an abnormal karyotype are the prime candidates for allogeneic transplantation. Experimental peripheral blood models that may reflect the degree of marrow fibrosis, such as the serum procollagen 3 peptide assay, have been used to determine if they are more informative of patient status than a single, random bone marrow sampling. Marrow fibrosis may be patchy, and thus a marrow biopsy alone without other data about marrow function may be misleading. Considerable long-term success in eradicating fibrosis and restoring normal cytogenetics, normal bone marrow morphology, and normal complete blood cell counts through transplantation has been reported. Many questions remain to be answered, however, before the appropriate role of hematopoietic stem cell transplantation in the setting of both spent-phase PV and IF can be determined.


Subject(s)
Hematopoietic Stem Cell Transplantation , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/therapy , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Polycythemia Vera/complications , Polycythemia Vera/mortality , Polycythemia Vera/therapy , Primary Myelofibrosis/etiology , Primary Myelofibrosis/therapy , Risk Factors , Survival Rate , Thrombocythemia, Essential/complications , Thrombocythemia, Essential/mortality
6.
Semin Hematol ; 41(2 Suppl 3): 18-22, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15190519

ABSTRACT

Myeloproliferative disorders (MPDs) are chronic conditions that require long-term treatment. MPDs have been considered neoplastic disorders that have a propensity to transform to acute leukemia or myelodysplastic syndrome. One of the MPDs, chronic myeloid leukemia (CML), has the distinct cytogenetic abnormality of the Philadelphia chromosome. In CML, transformation to acute leukemia is a common occurrence, with the timeline measured in years rather than decades. With the other three disorders from this group, polycythemia vera (PV), essential thrombocythemia (ET), and idiopathic myelofibrosis (IMF), transformation to acute leukemia is less frequently seen and, although variable, can take a longer period of time. The choice of therapy used for MPDs may contribute to leukemic transformation; chlorambucil, busulfan, and radiophosphorus are all examples of therapy that have been shown to be leukemogenic. Several studies have suggested the potential leukemogenicity of hydroxyurea (HU), but this remains controversial because no randomized studies have been conducted. The two newest agents used for MPDs, interferon-alpha and anagrelide, have both been studied for efficacy, but their influence on the potential for leukemic transformation has not been well studied to date.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Busulfan/therapeutic use , Chlorambucil/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/chemically induced , Myelodysplastic Syndromes/chemically induced , Myeloproliferative Disorders/drug therapy , Radiopharmaceuticals/therapeutic use , Antineoplastic Agents, Alkylating/adverse effects , Busulfan/adverse effects , Chlorambucil/adverse effects , Hydroxyurea/therapeutic use , Interferon-alpha/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Myelodysplastic Syndromes/etiology , Myeloproliferative Disorders/complications , Philadelphia Chromosome , Platelet Aggregation Inhibitors/therapeutic use , Quinazolines/therapeutic use , Radiopharmaceuticals/adverse effects
8.
Blood ; 105(11): 4508-15, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15705794

ABSTRACT

Idiopathic myelofibrosis (IM) is characterized by increased numbers of CD34(+) cells in the peripheral blood (PB). We explored the possible mechanisms underlying this abnormal trafficking of CD34(+) cells. Plasma levels of neutrophil elastase (NE), total and active matrix metalloproteinase 9 (MMP-9), and soluble vascular cell adhesion molecule-1 (sVCAM-1) were dramatically increased in IM. The absolute number of CD34(+) cells in the PB was correlated with the levels of sVCAM-1. Marked elevations of the levels of NE but not total and active MMP-9 as well as MMP-2 were detected in media conditioned by IM mononuclear cells (MNCs) as compared with that of healthy volunteers. IM MNC-conditioned media, however, was shown by zymographic analysis to contain increased gelatinolytic activity corresponding to the molecular weight of MMP-9. IM MNC-conditioned media also exhibited a greater ability to cleave VCAM-1 and c-kit in vitro, consistent with the biologic actions of NE. In addition, the increased ability of IM PB CD34(+) cells to migrate through a reconstituted basement membrane was diminished by several inhibitors of MMP-9 activity, indicating that these cells express increased levels of this MMP. These data indicate that a proteolytic environment exists in IM which might result in the sustained mobilization of CD34(+) cells.


Subject(s)
Cell Movement , Hematopoietic Stem Cells/pathology , Peptide Hydrolases/metabolism , Primary Myelofibrosis/enzymology , Antigens, CD34 , Blood Cells/pathology , Culture Media, Conditioned/chemistry , Humans , Leukocyte Elastase/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Proteins/metabolism , Primary Myelofibrosis/blood , Primary Myelofibrosis/etiology , Primary Myelofibrosis/pathology , Vascular Cell Adhesion Molecule-1/metabolism
9.
Blood ; 105(4): 1699-705, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15471948

ABSTRACT

Idiopathic myelofibrosis (IM) is characterized by the constitutive mobilization of CD34(+) cells. IM peripheral blood (PB) CD34(+) cells had a reduced cloning efficiency and a lower frequency of cobblestone areas compared with normal granulocyte colony-stimulating factor (G-CSF)-mobilized PB CD34(+) cells. IM CD34(+) cells engrafted nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice, demonstrating that they contain bone marrow (BM)-repopulating cells. G-CSF-mobilized CD34(+) cells produced multiple hematopoietic lineages within the NOD/SCID mice with a predominance of CD19(+) cells. By contrast, IM CD34(+) cells produced predominantly CD33(+) cells, increased numbers of CD41(+) cells, but fewer CD19(+) cells. Transcriptional clonality assays of the engrafted human IM cells demonstrated their clonal origin. CD34(+) cells from one patient isolated prior to leukemic transformation were capable of generating acute leukemia in NOD/SCID mice. The engrafted human cells exhibited the same abnormal karyotype as primary cells in a portion of the population. These findings demonstrate that BM-repopulating cells and more differentiated progenitor cells are constitutively mobilized into the PB in IM, and that their differentiation program is abnormal. In addition, the NOD/SCID model may be useful in gaining an understanding of the events occurring during the transition of IM to acute leukemia.


Subject(s)
Bone Marrow Cells/pathology , Cell Movement , Hematopoiesis , Primary Myelofibrosis/blood , Primary Myelofibrosis/pathology , Acute Disease , Animals , Antigens, CD34/biosynthesis , Bone Marrow Cells/immunology , Clone Cells , Female , Graft Survival/genetics , Graft Survival/immunology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Immunophenotyping , Leukemia/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Primary Myelofibrosis/genetics
10.
Biol Blood Marrow Transplant ; 10(11): 741-2, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15505604

ABSTRACT

Cord blood transplantation has been used extensively in the allogeneic setting for acquired and genetic disorders of hematopoiesis. There is less experience in the utility of autologous cord blood transplantation, and there is great controversy about the role of autologous cord blood collection and storage. We report on the successful use of autologous cord blood transplantation for the treatment of severe aplastic anemia following fulminant hepatic failure and living related liver transplantation.


Subject(s)
Anemia, Aplastic/therapy , Cord Blood Stem Cell Transplantation , Adult , Anemia, Aplastic/etiology , Child , Child, Preschool , Humans , Infant , Liver Failure, Acute/complications , Liver Failure, Acute/therapy , Liver Transplantation/adverse effects , Male , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL