Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Oncologist ; 28(12): 1034-1048, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-37774394

ABSTRACT

Colorectal cancer is the third most common cancer worldwide, and incidence is rising in younger individuals. Anti-EGFR antibodies, including cetuximab and panitumumab, have been incorporated into standard-of-care practice for patients with advanced disease. Herein, we review the molecular characteristics of these agents and the trials that lead to their approvals. Further, we discuss clinical implications of data regarding biomarkers that dictate treatment selection, different dosing strategies, and side effect management. Finally, we look towards the future and describe contexts in which these agents are currently being investigated clinically with a focus on combinations with MAPK-targeted therapies and immunotherapy. Overall, this review provides historical context, current clinical usage, and future directions for anti-EGFR antibodies in advanced colorectal cancer.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Humans , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , ErbB Receptors/genetics , ErbB Receptors/therapeutic use , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy
2.
Future Oncol ; 18(29): 3299-3310, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36066242

ABSTRACT

Aim: To examine characteristics of and treatment duration and real-world overall survival (rwOS) in patients receiving cetuximab as second-line (2L) or third-line (3L) treatment for metastatic colorectal cancer. Materials & methods: This was a retrospective study of 1096 and 684 patients in 2L and 3L cohorts, respectively. Results: The most common cetuximab-based regimens were cetuximab + folinic acid, fluorouracil and irinotecan (2L: 44%; 3L: 32%) and cetuximab + irinotecan (2L: 28%; 3L: 35%). Kaplan-Meier survival estimates and stepwise Cox regression model analysis demonstrated median treatment duration and rwOS of 3.7 and 14.4 months, respectively, in patients receiving treatment in the 2L cohort. In the 3L cohort, treatment duration was 3.3 months and rwOS was 12.0 months. Conclusion: This large real-world study provides evidence of rwOS in patients with metastatic colorectal cancer receiving cetuximab-based regimens as 2L or 3L treatment.


In this retrospective study, the authors examined baseline characteristics of and treatment duration and real-world overall survival (rwOS) in 1096 and 684 patients with metastatic colorectal cancer receiving cetuximab as second-line (2L) and third-line (3L) treatment, respectively. The most common cetuximab-based regimens were cetuximab + folinic acid, fluorouracil and irinotecan (2L: 44%; 3L: 32%) and cetuximab + irinotecan (2L: 28%; 3L: 35%). Median treatment duration and rwOS were 3.7 and 14.4 months, respectively, in patients receiving treatment in the 2L cohort. In the 3L cohort, median treatment duration was 3.3 months and rwOS was 12.0 months. This large real-world study provides evidence of rwOS in patients with metastatic colorectal cancer receiving cetuximab-based regimens as 2L or 3L treatment.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Rectal Neoplasms , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Camptothecin/therapeutic use , Cetuximab/adverse effects , Colonic Neoplasms/etiology , Colorectal Neoplasms/pathology , Fluorouracil/adverse effects , Humans , Irinotecan/therapeutic use , Leucovorin/adverse effects , Rectal Neoplasms/etiology , Retrospective Studies
3.
J Med Econ ; 24(1): 1164-1172, 2021.
Article in English | MEDLINE | ID: mdl-34529522

ABSTRACT

AIM: To compare the cost of biweekly regimens of first-line (1L) treatments of cetuximab-folinic acid, fluorouracil, and irinotecan (FOLFIRI) versus panitumumab-folinic acid, fluorouracil, and oxaliplatin (FOLFOX) in patients with Kirsten's rat sarcoma wild type (KRAS WT) metastatic colorectal cancer (mCRC) in the United States, across varying weights and body surface areas (BSAs). MATERIALS AND METHODS: Cost-minimization analysis (CMA) was performed to estimate per-patient cost differences of cetuximab-FOLFIRI versus panitumumab-FOLFOX. The CMA estimated the costs of RAS testing, premedication, drug acquisition, treating infusion reactions (IRs), supportive therapy, and biweekly administration of chemotherapy, cetuximab (500 mg/m2), and panitumumab (6 mg/kg) over 43 weeks (median progression-free survival). To calculate dose and cost, weight and height data were gathered from an electronic health record-derived de-identified database (n = 7,669; January 2013-October 2020). Base case analysis utilized mean weight/BSA of the overall cohort (82.04 kg/1.92 m2), and alternate scenarios were based on 88.18 kg/2.03 m2 (men, n = 4,477) and 73.43 kg/1.76 m2 (women, n = 3,192). RESULTS: For the base case, total treatment costs were $167,853 for cetuximab-FOLFIRI and $168,254 for panitumumab-FOLFOX; cost savings per patient receiving cetuximab-FOLFIRI was $400. Cost savings in alternate scenarios (men, $15,138; women, $15,004) resulted from lower drug acquisition costs for cetuximab (men, $14,833; women $14,854) and administration cost ($440) versus panitumumab. Cost savings of cetuximab-FOLFIRI in treating IR ($353) were similar across all scenarios. LIMITATIONS: With no head-to-head clinical trial data in the 1L setting, assumptions of similarity in efficacy and safety of cetuximab versus panitumumab were based on published network meta-analysis and the ASPECCT trial. This model did not consider a lifetime horizon. Costs of managing all adverse events (except IR) were not included. CONCLUSIONS: Biweekly cetuximab-FOLFIRI offers cost savings compared with panitumumab-FOLFOX for 1L therapy of patients with KRAS WT mCRC in the United States. These cost differences were observed for the overall population and across different BSA and weights for men and women.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins p21(ras) , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Costs and Cost Analysis , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Panitumumab/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , United States
4.
Int J Cancer ; 149(2): 403-408, 2021 07 15.
Article in English | MEDLINE | ID: mdl-33709428

ABSTRACT

Interleukin (IL)-10 has anti-inflammatory and CD8+ T-cell-stimulating properties. Pegilodecakin (pegylated recombinant human IL-10) induces intratumoral antigen-specific CD8 + T-cells and upregulates IFNγ and major histocompatibility complexes (MHC) I and II. Pegilodecakin has single-agent activity with manageable toxicity in advanced renal cell carcinama (aRCC) (data cutoff 24 March 2016). Pegilodecakin with pembrolizumab or nivolumab revealed clinical activity in aRCC (data cutoff 1 July 2018). Here, we report for the first time the results of pegilodecakin+ pazopanib, and final results for monotherapy and long-term follow-up with pegilodecakin + anti-programmed cell death 1 (anti-PD-1) inhibitors (data cutoff 19 February 2019). Phase 1/1b multi-cohort dose escalation IVY study enrolled 353 patients. Sixty-six patients with aRCC were treated with pegilodecakin alone or with pazopanib or anti-PD-1 inhibitor in cohorts A, G, H and I (data cutoff 19 February 2019). Primary endpoints included safety and tolerability. Secondary endpoint was tumor response by immune-related response criteria (irRC). Pegilodecakin plus nivolumab or pembrolizumab yielded median progression-free survival (mPFS) of 13.9 months and 6-month PFS probability of 60%, 76% 1-year overall survival (OS) probability and 61% 2-year OS probability. Pegilodecakin monotherapy produced mPFS of 1.8 months, 6-month PFS probability 25%, 1-year OS 50%, and 2-year OS 17%. Median OS was not reached in both combinations. Objective response rates (ORRs) were 33% with pazopanib and 43% with anti-PD-1. Most common Grade 3/4 treatment-related adverse events included anemia, thrombocytopenia and hypertriglyceridemia. In these heavily pretreated renal cell carcinama cohorts of IVY, pegilodecakin+anti-PD-1 inhibitor showed promising clinical activity. Safety profile of pegilodecakin alone and with anti-PD-1 inhibitors was consistent as previously reported.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Renal Cell/drug therapy , Indazoles/administration & dosage , Interleukin-10/administration & dosage , Kidney Neoplasms/drug therapy , Polyethylene Glycols/administration & dosage , Pyrimidines/administration & dosage , Sulfonamides/administration & dosage , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Administration Schedule , Female , Humans , Indazoles/therapeutic use , Interleukin-10/therapeutic use , Male , Middle Aged , Nivolumab/administration & dosage , Nivolumab/therapeutic use , Polyethylene Glycols/therapeutic use , Pyrimidines/therapeutic use , Sulfonamides/therapeutic use , Survival Analysis , Treatment Outcome , Young Adult
5.
Leuk Lymphoma ; 59(2): 416-422, 2018 02.
Article in English | MEDLINE | ID: mdl-28633612

ABSTRACT

Large granular lymphocytic leukemia (LGLL) represents a clonal/oligoclonal lymphoproliferation of cytotoxic T and natural killer cells often associated with STAT3 mutations. When symptomatic, due to mostly anemia and neutropenia, therapy choices are often empirically-based, because only few clinical trials and systematic studies have been performed. Incorporating new molecular and flow cytometry parameters, we identified 204 patients fulfilling uniform criteria for LGLL diagnoses and analyzed clinical course with median follow-up of 36 months, including responses to treatments. While selection of initial treatment was dictated by clinical features, the initial responses, as well as overall responses to methotrexate (MTX), cyclosporine (CsA), and cyclophosphamide (CTX), were similar at 40-50% across drugs. Sequential use of these drugs resulted in responses in most cases: only 10-20% required salvage therapies such as ATG, Campath, tofacitinib, splenectomy or abatacept. MTX yielded the most durable responses. STAT3-mutated patients required therapy more frequently and had better overall survival.


Subject(s)
Leukemia, Large Granular Lymphocytic/diagnosis , Leukemia, Large Granular Lymphocytic/mortality , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers , Female , High-Throughput Nucleotide Sequencing , Humans , Immunophenotyping , Leukemia, Large Granular Lymphocytic/genetics , Leukemia, Large Granular Lymphocytic/therapy , Male , Middle Aged , Mutation , Prognosis , Receptors, Antigen, T-Cell/genetics , STAT3 Transcription Factor/genetics , Survival Analysis , Symptom Assessment , Treatment Outcome
6.
Biol Blood Marrow Transplant ; 21(8): 1529-1531, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25937397

ABSTRACT

Brentuximab vedotin (BV), an antibody-drug conjugate that targets CD30, induces high response rates in CD30(+) lymphoid malignancies. It is unknown if BV use affects procurement of autologous CD34(+) stem cells and hematopoietic engraftment after autologous stem cell transplantation (ASCT). We examined 42 patients treated with BV before mobilization. Median times from diagnosis to transplantation, from initial BV treatment to transplantation, and from last BV treatment to stem cell collection were 21 months (range, 10 to 210), 5 months (range, 1.5 to 16.8), and 30 days (range, 2 to 280), respectively. Mobilization was successful on the first attempt in 38 patients (90.4%). The median number of infused CD34(+) cells was 5.46 × 10(6)/kg (range, 1.65 to 54.78 × 10(6)/kg). The median times to neutrophil and platelet engraftment were 10 (range, 9 to 13), and 10.5 days (range, 7 to 35), respectively. BV before high-dose chemotherapy-ASCT did not adversely affect peripheral blood stem cell mobilization and subsequent engraftment in a cohort of heavily pretreated patients with CD30(+) lymphomas.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Hodgkin Disease/therapy , Immunoconjugates/therapeutic use , Ki-1 Antigen/metabolism , Stem Cells/metabolism , Transplantation Conditioning/methods , Adolescent , Adult , Aged , Brentuximab Vedotin , Female , Humans , Immunoconjugates/administration & dosage , Male , Middle Aged , Retrospective Studies , Young Adult
7.
Blood ; 122(25): 4077-85, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24149287

ABSTRACT

New massively parallel sequencing technology enables, through deep sequencing of rearranged T-cell receptor (TCR) Vß complementarity-determining region 3 (CDR3) regions, a previously inaccessible level of TCR repertoire analysis. The CDR3 repertoire diversity reflects clonal composition, the potential antigenic recognition spectrum, and the quantity of available T-cell responses. In this context, T-large granular lymphocyte (T-LGL) leukemia is a chronic clonal lymphoproliferation of cytotoxic T cells often associated with autoimmune diseases and various cytopenias. Using CD8(+) T-LGL leukemia as a model disease, we set out to evaluate and compare the TCR deep-sequencing spectra of both patients and healthy controls to better understand how TCR deep sequencing could be used in the diagnosis and monitoring of not only T-LGL leukemia but also reactive processes such as autoimmune disease and infection. Our data demonstrate, with high resolution, significantly decreased diversity of the T-cell repertoire in CD8(+) T-LGL leukemia and suggest that many T-LGL clonotypes may be private to the disease and may not be present in the general public, even at the basal level.


Subject(s)
CD8-Positive T-Lymphocytes , Complementarity Determining Regions/genetics , Leukemia, Large Granular Lymphocytic/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Leukemia, Large Granular Lymphocytic/pathology , Male , Middle Aged
8.
Blood ; 122(14): 2453-9, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23926297

ABSTRACT

Large granular lymphocyte leukemia (LGL) is often associated with immune cytopenias and can cooccur in the context of aplastic anemia (AA) and myelodysplastic syndromes (MDS). We took advantage of the recent description of signal transducer and activator of transcription 3 (STAT3) mutations in LGL clonal expansions to test, using sensitive methods, for the presence of these mutations in a large cohort of 367 MDS and 140 AA cases. STAT3 clones can be found not only in known LGL concomitant cases, but in a small proportion of unsuspected ones (7% AA and 2.5% MDS). In STAT3-mutated AA patients, an interesting trend toward better responses of immunosuppressive therapy and an association with the presence of human leukocyte antigen-DR15 were found. MDSs harboring a STAT3 mutant clone showed a lower degree of bone marrow cellularity and a higher frequency of developing chromosome 7 abnormalities. STAT3-mutant LGL clones may facilitate a persistently dysregulated autoimmune activation, responsible for the primary induction of bone marrow failure in a subset of AA and MDS patients.


Subject(s)
Anemia, Aplastic/genetics , Leukemia, Large Granular Lymphocytic/genetics , Mutation , Myelodysplastic Syndromes/genetics , STAT3 Transcription Factor/genetics , Adult , Anemia, Aplastic/complications , Anemia, Aplastic/mortality , Cell Separation , Female , Flow Cytometry , Humans , Kaplan-Meier Estimate , Leukemia, Large Granular Lymphocytic/complications , Leukemia, Large Granular Lymphocytic/mortality , Male , Middle Aged , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/mortality , Proportional Hazards Models , Reverse Transcriptase Polymerase Chain Reaction
9.
Haematologica ; 98(1): 107-13, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22773603

ABSTRACT

Chronic myelomonocytic leukemia is a heterogeneous disease with multifactorial molecular pathogenesis. Various recurrent somatic mutations have been detected alone or in combination in chronic myelomonocytic leukemia. Recently, recurrent mutations in spliceosomal genes have been discovered. We investigated the contribution of U2AF1, SRSF2 and SF3B1 mutations in the pathogenesis of chronic myelomonocytic leukemia and closely related diseases. We genotyped a cohort of patients with chronic myelomonocytic leukemia, secondary acute myeloid leukemia derived from chronic myelomonocytic leukemia and juvenile myelomonocytic leukemia for somatic mutations in U2AF1, SRSF2, SF3B1 and in the other 12 most frequently affected genes in these conditions. Chromosomal abnormalities were assessed by nucleotide polymorphism array-based karyotyping. The presence of molecular lesions was correlated with clinical endpoints. Mutations in SRSF2, U2AF1 and SF3B1 were found in 32%, 13% and 6% of cases of chronic myelomonocytic leukemia, secondary acute myeloid leukemia derived from chronic myelomonocytic leukemia and juvenile myelomonocytic leukemia, respectively. Spliceosomal genes were affected in various combinations with other mutations, including TET2, ASXL1, CBL, EZH2, RAS, IDH1/2, DNMT3A, TP53, UTX and RUNX1. Worse overall survival was associated with mutations in U2AF1 (P=0.047) and DNMT3A (P=0.015). RAS mutations had an impact on overall survival in secondary acute myeloid leukemia (P=0.0456). By comparison, our screening of juvenile myelomonocytic leukemia cases showed mutations in ASXL1 (4%), CBL (10%), and RAS (6%) but not in IDH1/2, TET2, EZH2, DNMT3A or the three spliceosomal genes. SRSF2 and U2AF1 along with TET2 (48%) and ASXL1 (38%) are frequently affected by somatic mutations in chronic myelomonocytic leukemia, quite distinctly from the profile seen in juvenile myelomonocytic leukemia. Our data also suggest that spliceosomal mutations are of ancestral origin.


Subject(s)
Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Spliceosomes/genetics , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Infant , Leukemia, Myelomonocytic, Chronic/diagnosis , Leukemia, Myelomonocytic, Juvenile/diagnosis , Male , Middle Aged
10.
SAGE Open Med Case Rep ; 1: 2050313X13498709, 2013.
Article in English | MEDLINE | ID: mdl-27489629

ABSTRACT

INTRODUCTION: Autoimmune myelofibrosis is an uncommon hematologic disease characterized by anemia, bone marrow myelofibrosis, and an autoimmune feature. Myelofibrosis is often associated with other conditions, including infections, nutritional/endocrine dysfunction, toxin/drug exposure, and connective tissue diseases, including scleroderma and systemic lupus erythematosus. Absence of clonal markers (JAK2) and heterogeneity of the symptoms often complicate the diagnosis. CASE PRESENTATION: Here, we present two cases of systemic lupus erythematosus-induced autoimmune myelofibrosis. The first case is of a 36-year-old African American female with diagnosis of systemic lupus erythematosus at the age of 12 years. The second patient is a 44-year-old African American male with family history of systemic lupus erythematosus who developed anemia and constitutional symptoms later on. Both patients showed hypercellularity and fibrotic changes of the bone marrow. Moreover, mutational analysis showed that both patients were wild type for JAK2 (V617F and exon 12) and MPL (exon 10). CONCLUSIONS: These two cases illustrate that anemic patients with fibrotic changes in the bone marrow without other clinicopathologic features associated with primary myelofibrosis in the presence of clinical manifestations and history of an autoimmune disease should suggest an autoimmune myelofibrosis. These cases demonstrate that a good clinical history combined with molecular technologies and pathomorphologic criteria are helpful in distinguishing between primary myelofibrosis and a nonclonal myelofibrosis from an associated condition.

11.
Blood ; 120(15): 3048-57, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-22859607

ABSTRACT

Chronic lymphoproliferative disorders of natural killer cells (CLPD-NKs) and T-cell large granular lymphocytic leukemias (T-LGLs) are clonal lymphoproliferations arising from either natural killer cells or cytotoxic T lymphocytes (CTLs). We have investigated for distribution and functional significance of mutations in 50 CLPD-NKs and 120 T-LGL patients by direct sequencing, allele-specific PCR, and microarray analysis. STAT3 gene mutations are present in both T and NK diseases: approximately one-third of patients with each type of disorder convey these mutations. Mutations were found in exons 21 and 20, encoding the Src homology 2 domain. Patients with mutations are characterized by symptomatic disease (75%), history of multiple treatments, and a specific pattern of STAT3 activation and gene deregulation, including increased expression of genes activated by STAT3. Many of these features are also found in patients with wild-type STAT3, indicating that other mechanisms of STAT3 activation can be operative in these chronic lymphoproliferative disorders. Treatment with STAT3 inhibitors, both in wild-type and mutant cases, resulted in accelerated apoptosis. STAT3 mutations are frequent in large granular lymphocytes suggesting a similar molecular dysregulation in malignant chronic expansions of NK and CTL origin. STAT3 mutations may distinguish truly malignant lymphoproliferations involving T and NK cells from reactive expansions.


Subject(s)
Killer Cells, Natural/pathology , Leukemia, Large Granular Lymphocytic/genetics , Leukemia, Large Granular Lymphocytic/pathology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/pathology , Mutation/genetics , STAT3 Transcription Factor/genetics , Adult , Aged , Aged, 80 and over , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression Profiling , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , Young Adult
12.
Blood ; 120(25): 4945-51, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-22915641

ABSTRACT

Lenalidomide and azacitidine each have activity in myelodysplastic syndromes (MDS) patients, where both microenvironment and cell-regulatory mechanisms contribute to disease pathogenesis. The objective of this multicenter, phase 2 expansion trial was to determine the efficacy and safety of combination therapy with azacitidine (75 mg/m(2)/d for 5 days) and lenalidomide (10 mg/d for 21 days; 28-day cycle) in patients with higher-risk MDS. Among 36 patients enrolled (18 phase 1, 18 phase 2), median age was 68 years (range, 47-78 years) and follow-up was 12 months (range, 3-55 years). IPSS categories included intermediate-1 (n = 5 patients with excess blasts), intermediate-2 (20), and high (11). Common grade 3/4 nonhematologic adverse events included febrile neutropenia (22% of patients), other infection (11%), pulmonary (11%), cardiac (11%), constitutional (11%), and dermatologic (11%). The overall response rate (per modified MDS International Working Group criteria) was 72%: 16 patients (44%) achieved a complete response (CR), and 10 (28%) had hematologic improvement. Median CR duration was 17+ months (range, 3-39+); median overall survival was 37+ months (range, 7-55+) for CR patients, and 13.6 months for the entire cohort (range, 3-55). TET2/DNMT3A/IDH1/2 mutational status was associated with response in a limited number of patients. The lenalidomide/azacitidine combination is well-tolerated and highly active in treating greater-risk MDS.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/therapeutic use , Myelodysplastic Syndromes/drug therapy , Thalidomide/analogs & derivatives , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Azacitidine/administration & dosage , Azacitidine/adverse effects , Cohort Studies , DNA Mutational Analysis , Female , Humans , Karyotyping , Lenalidomide , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/therapeutic use , Treatment Outcome
13.
J Hematol Oncol ; 5: 4, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22390313

ABSTRACT

BACKGROUND: While lenalidomide (LEN) shows high efficacy in myelodysplastic syndromes (MDS) with del[5q], responses can be also seen in patients presenting without del[5q]. We hypothesized that improved detection of chromosomal abnormalities with new karyotyping tools may better predict response to LEN. DESIGN AND METHODS: We have studied clinical, molecular and cytogenetic features of 42 patients with MDS, myeloproliferative neoplasms (MPN), MDS/MPN overlap syndromes and secondary acute myeloid leukemia (sAML) without del[5q] by metaphase cytogenetics (MC) who underwent therapy with LEN. RESULTS: Fluorescence in situ hybridization (FISH) or single nucleotide polymorphism array (SNP-A)-based karyotyping marginally increased the diagnostic yield over MC, detecting 2/42 (4.8%) additional cases with del[5q], one of whom were responded to LEN. Responses were more often observed in patients with a normal karyotype by MC (60% vs abnormal MC; 17%, p = .08) and those with gain of chromosome 8 material by either of all 3 karyotyping methods (83% vs all other chromosomal abnormalities; 44% p = .11). However, 5 out of those 6 patients received combined LEN/AZA therapy and it may also suggest those with gain of chromosome 8 material respond well to AZA. The addition of FISH or SNP-A did not improve the predictive value of normal cytogenetics by MC. Mutational analysis of TET2, UTX, CBL, EZH2, ASXL1, TP53, RAS, IDH1/2, and DNMT-3A was performed on 21 of 41 patients, and revealed 13 mutations in 11 patients, but did not show any molecular markers of responsiveness to LEN. CONCLUSIONS: Normal karyotype and gain of chromosome 8 material was predictive of response to LEN in non-del[5q] patients with myeloid malignancies.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/genetics , Thalidomide/analogs & derivatives , Aged , Aged, 80 and over , Chromosome Aberrations , DNA Methyltransferase 3A , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Lenalidomide , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Mutation/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/pathology , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/pathology , Polymorphism, Single Nucleotide/genetics , Prognosis , Thalidomide/therapeutic use
14.
Blood ; 119(14): 3203-10, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-22323480

ABSTRACT

Myelodysplastic syndromes (MDSs) are chronic and often progressive myeloid neoplasms associated with remarkable heterogeneity in the histomorphology and clinical course. Various somatic mutations are involved in the pathogenesis of MDS. Recently, mutations in a gene encoding a spliceosomal protein, SF3B1, were discovered in a distinct form of MDS with ring sideroblasts. Whole exome sequencing of 15 patients with myeloid neoplasms was performed, and somatic mutations in spliceosomal genes were identified. Sanger sequencing of 310 patients was performed to assess phenotype/genotype associations. To determine the functional effect of spliceosomal mutations, we evaluated pre-mRNA splicing profiles by RNA deep sequencing. We identified additional somatic mutations in spliceosomal genes, including SF3B1, U2AF1, and SRSF2. These mutations alter pre-mRNA splicing patterns. SF3B1 mutations are prevalent in low-risk MDS with ring sideroblasts, whereas U2AF1 and SRSF2 mutations are frequent in chronic myelomonocytic leukemia and advanced forms of MDS. SF3B1 mutations are associated with a favorable prognosis, whereas U2AF1 and SRSF2 mutations are predictive for shorter survival. Mutations affecting spliceosomal genes that result in defective splicing are a new leukemogenic pathway. Spliceosomal genes are probably tumor suppressors, and their mutations may constitute diagnostic biomarkers that could potentially serve as therapeutic targets.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mutation , Nuclear Proteins/genetics , Phosphoproteins/genetics , RNA Splicing/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Ribonucleoproteins/genetics , Base Sequence , Female , Genetic Association Studies , Humans , Leukemia, Myeloid/diagnosis , Leukemia, Myeloid/genetics , Leukemia, Myeloid/mortality , Male , Mutation Rate , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Prognosis , RNA Splicing Factors , Ribonucleoprotein, U2 Small Nuclear/metabolism , Ribonucleoproteins/metabolism , Sequence Alignment , Serine-Arginine Splicing Factors , Spliceosomes/genetics , Spliceosomes/metabolism , Splicing Factor U2AF
15.
Article in English | MEDLINE | ID: mdl-22160018

ABSTRACT

Current immunosuppressive treatment (IST) induces remissions in 50%-70% of patients with aplastic anemia (AA) and result in excellent long-term survival. In recent years, the survival of refractory patients has also improved. Apart from relapse and refractoriness to IST, evolution of clonal diseases, including paroxysmal nocturnal hemoglobinuria and myelodysplastic syndrome (MDS), are the most serious long-term complications and constitute a strong argument for definitive therapy with BM transplantation if possible. Consequently, the detection of diagnostic chromosomal abnormalities (mostly monosomy 7) is of great clinical importance. Newer whole-genome scanning technologies such as single nucleotide polymorphism (SNP) array-based karyotyping may be a helpful diagnostic test for the detection of chromosomal defects in AA due to its precision/resolution and lack of reliance on cell division.


Subject(s)
Anemia, Aplastic/genetics , Anemia, Aplastic/pathology , Clonal Evolution/genetics , Anemia, Aplastic/diagnosis , Anemia, Aplastic/therapy , Humans , Myelodysplastic Syndromes/diagnosis
16.
Haematologica ; 96(9): 1269-75, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21606164

ABSTRACT

BACKGROUND: A combination of horse anti-thymocyte globulin and cyclosporine produces responses in 60-70% of patients with severe aplastic anemia. We performed a phase II study of rabbit anti-thymocyte globulin and cyclosporine as first-line therapy for severe aplastic anemia. DESIGN AND METHODS: Twenty patients with severe aplastic anemia treated with rabbit anti-thymocyte globulin were compared to 67 historical control cases with matched clinical characteristics treated with horse anti-thymocyte globulin. RESULTS: Response rates at 3, 6 and 12 months were similar for patients treated with rabbit anti-thymocyte globulin or horse anti-thymocyte globulin: 40% versus 55% (P=0.43), 45% versus 58% (P=0.44) and 50% versus 58% (P=0.61), respectively. No differences in early mortality rates or overall survival were observed. We then performed multivariable analyses of response at 6 months and overall survival and identified the presence of a paroxysmal nocturnal hemoglobinuria clone (P=0.01) and a pretreatment absolute reticulocyte count greater than 30×10(9)/L (P=0.007) as independent predictors of response and younger age (P=0.003), higher pretreatment absolute neutrophil (P=0.02) and absolute lymphocyte counts (P=0.03) as independent predictors of overall survival. None of the immunogenetic polymorphisms studied was predictive of response to immunosupressive therapy. CONCLUSIONS: Despite reports suggesting differences in biological activity of different anti-thymocyte globulin preparations, rabbit and horse anti-thymocyte globulin appear to have a similar efficacy for up-front treatment of severe aplastic anemia. Clinicaltrial.gov: NCT01231841).


Subject(s)
Anemia, Aplastic/drug therapy , Antilymphocyte Serum/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Anemia, Aplastic/genetics , Anemia, Aplastic/mortality , Animals , Antilymphocyte Serum/administration & dosage , Antilymphocyte Serum/adverse effects , Child , Child, Preschool , Cyclosporine/therapeutic use , Cytokines/genetics , Female , Horses , Humans , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Polymorphism, Genetic , Rabbits , Recurrence , Survival Analysis , Treatment Outcome , Young Adult
17.
Blood ; 117(25): 6876-84, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21527527

ABSTRACT

In aplastic anemia (AA), contraction of the stem cell pool may result in oligoclonality, while in myelodysplastic syndromes (MDS) a single hematopoietic clone often characterized by chromosomal aberrations expands and outcompetes normal stem cells. We analyzed patients with AA (N = 93) and hypocellular MDS (hMDS, N = 24) using single nucleotide polymorphism arrays (SNP-A) complementing routine cytogenetics. We hypothesized that clinically important cryptic clonal aberrations may exist in some patients with BM failure. Combined metaphase and SNP-A karyotyping improved detection of chromosomal lesions: 19% and 54% of AA and hMDS cases harbored clonal abnormalities including copy-neutral loss of heterozygosity (UPD, 7%). Remarkably, lesions involving the HLA locus suggestive of clonal immune escape were found in 3 of 93 patients with AA. In hMDS, additional clonal lesions were detected in 5 (36%) of 14 patients with normal/noninformative routine cytogenetics. In a subset of AA patients studied at presentation, persistent chromosomal genomic lesions were found in 10 of 33, suggesting that the initial diagnosis may have been hMDS. Similarly, using SNP-A, earlier clonal evolution was found in 4 of 7 AA patients followed serially. In sum, our results indicate that SNP-A identify cryptic clonal genomic aberrations in AA and hMDS leading to improved distinction of these disease entities.


Subject(s)
Anemia, Aplastic/genetics , Myelodysplastic Syndromes/genetics , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Adult , Aged , Aged, 80 and over , Chromosome Aberrations , Cytogenetic Analysis , Female , Humans , Karyotyping/methods , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods
18.
Cancer ; 117(6): 1253-61, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-20960521

ABSTRACT

BACKGROUND: Higher-risk myelodysplastic syndromes (MDS) are similar pathobiologically to acute myeloid leukemia (AML), particularly in older adults. AML therapies thus may have activity in MDS. In the current study, phase 2 study data of arsenic trioxide (ATO) and gemtuzumab ozogamicin (GO) in CD33-positive patients with MDS and secondary AML (sAML) were presented. METHODS: Between June 2004 and February 2006, 30 patients with higher-risk MDS or sAML received ATO (at a dose of 0.25 mg/kg intravenously for 5 days during Week 1, then twice weekly during Weeks 2-12) and GO (at a dose of 3 mg/m(2) on Day 8) for 1 or 2 cycles of 12 weeks each. The primary endpoint was response as per MDS or AML International Working Group (IWG) criteria. Adverse events were collected throughout treatment. Patients were followed for a minimum of 3 years for survival. RESULTS: The median patient age was 69 years. A total of 18 patients had MDS, 12 had sAML, and 19 had been previously treated. Seventeen patients (57%) completed ≥1 cycle, and 7 patients (23%) completed 2 cycles. IWG responses occurred in 9 patients (30%) according to IWG MDS criteria (including 2 of 7 patients who failed hypomethylating agents) and 3 of 12 AML patients (25%) according to IWG AML criteria. Grade 3/4 (according to National Cancer Institute Common Toxicity Criteria [version 3.0]) thrombocytopenia occurred in 47% of patients, neutropenia in 63%, and anemia in 37% of patients. The median overall survival was 9.7 months (28.6 months in responders and 7.6 months in nonresponders; P <.001). Patients who completed 2 cycles of therapy spent a median of 13 days in the hospital. CONCLUSIONS: Combination therapy with ATO and GO was found to have acceptable response rates and toxicity, and may be a viable treatment option to standard induction therapy, particularly for patients who fail therapy with hypomethylating agents.


Subject(s)
Aminoglycosides/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenicals/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Neoplasms, Second Primary/drug therapy , Oxides/administration & dosage , Adult , Aged , Aged, 80 and over , Aminoglycosides/adverse effects , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Arsenic Trioxide , Arsenicals/adverse effects , Drug Administration Schedule , Female , Gemtuzumab , Humans , Male , Middle Aged , Myelodysplastic Syndromes/pathology , Oxides/adverse effects , Salvage Therapy , Treatment Outcome
19.
Am J Hematol ; 86(1): 102-3, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21080340

ABSTRACT

Lenalidomide and azacitidine are active in MDS patients, and may complement each other by targeting the bone marrow microenvironment and the malignant clone. A recent Phase I trial testing the lenalidomide and azacitidine combination yielded encouraging results; however, lenalidomide's contribution was unclear. In this study, 18 higher-risk MDS patients were treated with the combination for seven cycles, after which lenalidomide was discontinued in eight patients who achieved a complete response, with azacitidine monotherapy continuing until disease progression. We report on three patients who relapsed on monotherapy with excess blasts at 12, 19, and 24 months, in whom lenalidomide was then resumed in combination with azacitidine. Each patient, one with normal cytogenetics at relapse; one with a 18 abnormality; and one with del(4q25), recaptured a complete response that was sustained for 5, 7, and 7+ months. We conclude that the addition of lenalidomide to azacitidine provides additional clinical benefit over azacitidine monotherapy.


Subject(s)
Anemia, Refractory, with Excess of Blasts/drug therapy , Azacitidine/therapeutic use , Thalidomide/analogs & derivatives , Aged , Humans , Lenalidomide , Male , Middle Aged , Risk Factors , Thalidomide/therapeutic use
20.
J Clin Oncol ; 28(13): 2253-8, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20354132

ABSTRACT

PURPOSE: Lenalidomide and azacitidine are active in patients with lower- and higher-risk myelodysplastic syndromes (MDS). These agents may complement each other by targeting both the bone marrow microenvironment and hypomethylating action on the malignant clone. PATIENTS AND METHODS: This phase I trial explored the safety of combination therapy in patients with higher-risk MDS. Response and characterization of molecular and methylation status of responders were secondary objectives. Patients were enrolled using a 3 + 3 dose escalation. Cycles lasted 28 days, and patients received a maximum of seven cycles. RESULTS: Of 18 patients enrolled, median age was 68 years (range, 52 to 78 years), interval from diagnosis was 5 weeks (range, 2 to 106 weeks), and follow-up was 7 months (range, 1 to 26 months). International Prognostic Scoring System categories were intermediate 1 (n = 2), intermediate 2 (n = 10), and high (n = 6). No dose-limiting toxicities occurred, and a maximum-tolerated dose was not reached. Grades 3 to 4 nonhematologic toxicities (> 1) included febrile neutropenia (n = 5), cardiac (n = 2), and CNS hemorrhage (n = 2). Median absolute neutrophil count decrease was 26%, and platelet decrease was 1% (mean, 24%). The overall response rate was 67%: eight patients (44%) had a complete response (CR); three patients (17%) had hematologic improvement; one patient (6%) had marrow CR. Patients achieving CR were more likely to have normal cytogenetics and lower methylation levels. CONCLUSION: The combination of lenalidomide and azacitidine is well tolerated with encouraging clinical activity. The go-forward dose is azacitidine 75 mg/m(2) on days 1 through 5 and lenalidomide 10 mg on days 1 through 21.


Subject(s)
Azacitidine/administration & dosage , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Myelodysplastic Syndromes/drug therapy , Precancerous Conditions/drug therapy , Thalidomide/analogs & derivatives , Aged , Azacitidine/adverse effects , Chromosome Aberrations , DNA Mutational Analysis , Drug Administration Schedule , Drug Therapy, Combination , Enzyme Inhibitors/adverse effects , Female , Humans , Karyotyping , Lenalidomide , Male , Middle Aged , Mutation , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/genetics , Precancerous Conditions/enzymology , Precancerous Conditions/genetics , Risk Assessment , Thalidomide/administration & dosage , Thalidomide/adverse effects , Time Factors , Treatment Outcome , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...