Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Oncotarget ; 13: 986-1002, 2022.
Article in English | MEDLINE | ID: mdl-36093297

ABSTRACT

Classical MCL (cMCL) constitutes 6-8% of all B cell NHL. Despite recent advances, MCL is incurable except with allogeneic stem cell transplant. Blastic mantle cell lymphoma (bMCL) is a rarer subtype of cMCL associated with an aggressive clinical course and poor treatment response, frequent relapse and poor outcomes. We treated 13 bMCL patients with combined epigenetic and immunotherapy treatment consisting of vorinostat, cladribine and rituximab (SCR). We report an increased OS greater than 40 months with several patients maintaining durable remissions without relapse for longer than 5 years. This is remarkably better then current treatment regimens which in bMCL range from 14.5-24 months with conventional chemotherapy regimens. We demonstrate that the G/A870 CCND1 polymorphism is predictive of blastic disease, nuclear localization of cyclinD1 and response to SCR therapy. The major resistance mechanisms to SCR therapy are loss of CD20 expression and evasion of treatment by sanctuary in the CNS. These data indicate that administration of epigenetic agents improves efficacy of anti-CD20 immunotherapies. This approach is promising in the treatment of MCL and potentially other previously treatment refractory cancers.


Subject(s)
Epigenesis, Genetic , Immunotherapy , Lymphoma, Mantle-Cell , Adult , Antigens, CD20/immunology , Cladribine , Humans , Immunologic Factors/therapeutic use , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/therapy , Rituximab/therapeutic use , Vorinostat/therapeutic use
2.
Ther Adv Hematol ; 13: 20406207221080743, 2022.
Article in English | MEDLINE | ID: mdl-35237397

ABSTRACT

The heterogeneity in disease pathology, the unpredictability in disease prognosis, and the variability in response to therapy make mantle cell lymphoma (MCL) a focus of novel therapeutic development. MCL is characterized by dysregulated expression of cyclin D1 through a chromosome t(11;14) translocation. MCL international prognostic index (MIPI), ki-67 proliferation index, and TP53 mutation status are currently utilized for prognostication. With advances in pharmacokinetic analysis and drug discovery, treatment strategy has evolved from chemotherapy to combination of targeted, epigenetic, and immune therapies. In this review, we discuss investigational and newly approved treatment approaches. In a short time, the US Food and Drug Administration (FDA) has approved five agents for the treatment of MCL: lenalidomide, an immunomodulatory agent; bortezomib, a proteasome inhibitor; and ibrutinib, acalabrutinib, and zanubrutinib, all Bruton kinase inhibitors. Epigenetic agents (e.g. cladribine and vorinostat), mammalian target of rapamycin (mTOR) inhibitors (e.g. temsirolimus and everolimus), and monoclonal antibodies and/or antibody-drug conjugates (e.g. obinutuzumab, polatuzumab, and ublituximab) are promising therapeutic agents currently under clinical trial investigation. Most recently, chimeric antigen receptor (CAR)-T cell therapy and bispecific T-cell engager (BiTE) therapy even open a new venue for MCL treatment. However, due to its intricate pathology nature and high relapse incidence, there are still unmet needs in developing optimal therapeutic strategies for both frontline and relapsed/refractory settings. The ultimate goal is to develop innovative personalized combination therapy approaches for the purpose of delivering precision medicine to cure this disease.

3.
Br J Haematol ; 186(6): 845-854, 2019 09.
Article in English | MEDLINE | ID: mdl-31177537

ABSTRACT

Altered DNA methylation and histone acetylation in lymphoma provided the rationale for using vorinostat (SAHA), cladribine and rituximab (SCR) in non-Hodgkin lymphomas (NHL) in this phase 1-2 study (NCT00764517). Treatment included cladribine 5 mg/m2 intravenously (IV) (days 1-5), rituximab 375 mg/m2 IV (weekly 4× for cycle 1 and 1×/month) and vorinostat orally once daily (days 1-14) every 28 days for up to six cycles. Phase 1 included relapsed patients (n = 10) in a standard 3 + 3 dose escalation design (vorinostat: 200, 300 and 400 mg). No dose-limiting toxicities were seen. The phase 2 dose for vorinostat was 400 mg po (days 1-14). The majority of phase 2 patients had mantle cell lymphoma (MCL) (n = 57; 39 previously untreated, 10 relapsed). The primary objective was objective response rate [complete response (CR) + partial response] which was 39% (7/18) in relapsed patients and 97% (38/39) with 80% (31/39) attaining a CR in previously untreated MCL. At a median follow-up of 42 months, median progression-free survival (PFS) and overall survival (OS) for relapsed NHL were 19·5 [95% confidence interval (CI): 2·0-33·0] and 25·0 (95% CI: 12·0-45·0) months respectively. Median PFS for previously untreated MCL was 84·0 months; OS could not be estimated. Toxicities were primarily haematological.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Lymphoma, B-Cell , Lymphoma, Mantle-Cell , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cladribine/administration & dosage , Cladribine/adverse effects , Disease-Free Survival , Female , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/mortality , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/mortality , Male , Middle Aged , Recurrence , Rituximab/administration & dosage , Rituximab/adverse effects , Survival Rate , Vorinostat/administration & dosage , Vorinostat/adverse effects
4.
Exp Hematol Oncol ; 8: 2, 2019.
Article in English | MEDLINE | ID: mdl-30733891

ABSTRACT

Mantle cell lymphoma is a relatively new recognized hematological malignant disease, comprising of 2.5-6% non-Hodgkin's lymphomas. The complexity of its clinical presentations (nodular pattern, diffuse pattern, and blastoid variant), variety in disease progression, and treatment response, make this disease a research focus to both experimental oncology and clinical oncology. Overexpression of cyclin D1 and chromosome t(11,14) translocation are the known molecular biomarkers of this disease. Mantle cell international prognostic index (MIPI), ki-67 proliferation index, and TP53 mutation are emerging as the prognostic biomarkers. Epigenetic profile variance and SOX11 gene expression profile correlate with treatment response. Over the years, the treatment strategy has been gradually evolving from combination chemotherapy to combination of targeted therapy, epigenetic modulation therapy, and immunotherapy. In a surprisingly short period of time, FDA specifically approved 4 drugs for treating mantle cell lymphoma: lenalidomide, an immunomodulatory agent; Bortezomib, a proteasome inhibitor; and Ibrutinib and acalabrutinib, both Bruton kinase inhibitors. Epigenetic agents (e.g. Cladribine and Vorinostat) and mTOR inhibitors (e.g. Temsirolimus and Everolimus) have been showing promising results in several clinical trials. However, treating aggressive variants of this disease that appear to be refractory/relapse to multiple lines of treatment, even after allogeneic stem cell transplant, is still a serious challenge. Developing a personalized, precise therapeutic strategy combining targeted therapy, immunotherapy, epigenetic modulating therapy, and cellular therapy is the direction of finding a curative therapy for this subgroup of patients.

5.
Exp Hematol ; 44(3): 157-60, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26802532

ABSTRACT

We recently reported that addition of epigenetic agents could overcome resistance of leukemic cells to monoclonal antibody-mediated anti-tumor effects in T-cell prolymphocytic leukemia. We also reported that epigenetic agents could induce expression of the CD30 gene, thus providing a therapeutic target for the antibody drug conjugate brentuximab vedotin. Here we discuss these findings and their generality to treatment of other hematologic and solid malignancies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Drug Resistance, Neoplasm , Epigenesis, Genetic/drug effects , Hematologic Neoplasms/therapy , Immunoconjugates/therapeutic use , Immunotherapy/methods , Animals , Brentuximab Vedotin , Hematologic Neoplasms/metabolism , Humans
6.
Sci Transl Med ; 7(293): 293ra102, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26109102

ABSTRACT

T cell prolymphocytic leukemia (T-PLL) is a rare, mature T cell neoplasm with distinct features and an aggressive clinical course. Early relapse and short overall survival are commonplace. Use of the monoclonal anti-CD52 antibody alemtuzumab has improved the rate of complete remission and duration of response to more than 50% and between 6 and 12 months, respectively. Despite this advance, without an allogeneic transplant, resistant relapse is inevitable. We report seven complete and one partial remission in eight patients receiving alemtuzumab and cladribine with or without a histone deacetylase inhibitor. These data show that administration of epigenetic agents can overcome alemtuzumab resistance. We also report epigenetically induced expression of the surface receptor protein CD30 in T-PLL. Subsequent treatment with the anti-CD30 antibody-drug conjugate brentuximab vedotin overcame organ-specific (skin) resistance to alemtuzumab. Our findings demonstrate activity of combination epigenetic and immunotherapy in the incurable illness T-PLL, particularly in the setting of previous alemtuzumab therapy.


Subject(s)
Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Leukemia, Prolymphocytic, T-Cell/genetics , Aged , Alemtuzumab , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brentuximab Vedotin , Cell Proliferation/drug effects , Chromatin/metabolism , Drug Resistance, Neoplasm/drug effects , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Leukemic/drug effects , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Ki-1 Antigen/metabolism , Leukemia, Prolymphocytic, T-Cell/blood , Leukemia, Prolymphocytic, T-Cell/drug therapy , Leukocyte Count , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , STAT5 Transcription Factor/genetics , Skin/drug effects , Skin/pathology , Treatment Outcome
7.
Mol Cancer Ther ; 13(12): 2784-92, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25319394

ABSTRACT

With an increasing number of clinical trials looking at combination therapies in cancer, potential drug-drug interactions require particular attention. One such instance is the treatment of CD30(+) tumors after previous vorinostat (SAHA; suberoylanilide hydroxyamic acid) failure with the anti-CD30 antibody-drug conjugate brentuximab vedotin. Using B-, T-, and natural killer (NK)-cell lines in vitro, we demonstrate that SAHA downregulates the expression of CD30 and lowers the efficacy of subsequent brentuximab vedotin treatment if baseline CD30 levels are reduced by 50% or more. Interestingly, low-dose SAHA treatment that maintained 50% or more of basal CD30 expression followed by subsequent treatment with brentuximab vedotin led to enhanced antitumor activity. The downregulation of CD30 was short lived upon SAHA removal, suggesting that allowing SAHA washout may circumvent any interactions with subsequent drug therapies. Our findings confirm the requirement of CD30 for brentuximab vedotin efficacy and suggest that combination treatment with SAHA in CD30(dim) tumors may decrease efficacy. Combination treatment in highly CD30(+) tumors, however, increases efficacy and warrants further consideration as a new treatment paradigm.


Subject(s)
Hydroxamic Acids/pharmacology , Immunoconjugates/pharmacology , Ki-1 Antigen/antagonists & inhibitors , Lymphocytes/drug effects , Lymphocytes/metabolism , Brentuximab Vedotin , Cell Line, Tumor , Cell Membrane/metabolism , Drug Antagonism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Ki-1 Antigen/genetics , Lymphocytes/immunology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Vorinostat
8.
Adv Exp Med Biol ; 779: 257-66, 2013.
Article in English | MEDLINE | ID: mdl-23288643

ABSTRACT

Epigenetics is the study of heritable changes in phenotype or gene expression caused by mechanisms other than changes in the underlying DNA sequence. Such changes can include DNA methylation or histone modifications which both serve to silence gene expression. This review describes a new development in pharmacology, epigenetic therapy, which attempts to correct these epigenetic changes for the treatment of mantle cell lymphoma (MCL) and other B cell malignancies for which no consensus on standard therapy exists. One class of drugs utilized are the histone deacetylase inhibitors, (HDACi) which result in the accumulation of acetylated histones. Hyperacetylation of histones and nonhistone proteins are postulated to mediate the anticancer effects of these drugs. Another class of epigenetic agents are hypomethylating agents, that can cause both DNA and histone hypomethylation. Epigenetic drugs may be useful in the treatment of cancer where hypermethylation of tumor suppressor genes is known to lead to silencing of these genes. The purine analog cladribine has been shown to have hypomethylating properties and has activity as a single agent or in combination with other therapies for mantle cell lymphoma. Epigenetic therapy with the DNA hypomethylating agent 5-aza-2-deoxycytidine can also cause restoration of cell surface expression of the CD20 protein and increase rituximab sensitivity in vitro. Combinations of epigenetic agents may act synergistically to further potentiate the efficacy of monoclonal antibodies like rituximab and ofatumumab and improve the treatment outcome in MCL.


Subject(s)
Epigenesis, Genetic , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/genetics , DNA Methylation , Humans , Neoplastic Stem Cells/drug effects , Tumor Microenvironment/drug effects
9.
Best Pract Res Clin Haematol ; 25(2): 143-52, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22687450

ABSTRACT

Mantle cell lymphoma (MCL) is a rare, aggressive subtype of B cell NHL for which there is no standard of care. It is characterized by the t(11;14) translocation, implicating cyclin D1 (CCND1) in its pathogenesis. Cyclin D1 is one of a family of 3 unlinked D type cyclin genes, CCND1, 2, 3. CCND1 is not expressed in normal B cells. Deregulated expression occurs as a result of juxtaposition of cis IgH enhancer elements, Eµ and 3' Cα, to the cyclin D1 gene. These enhancer elements and regions upstream of the CCND1 gene are hypomethylated on the translocated allele. Histones surrounding the translocation have shown hyperacetylation as well, a hallmark of transcriptionally active chromatin. The t(11;14) translocation is an epigenetic event, leading to cyclin D1 deregulated transcription. These findings provide the rationale for the use of epigenetic and targeted cyclin D1 therapies to overcome resistance and induce durable remissions in MCL.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclin D1/genetics , Epigenesis, Genetic/drug effects , Lymphoma, Mantle-Cell/drug therapy , Antineoplastic Agents/administration & dosage , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Cycle/drug effects , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 14 , Cyclin D1/antagonists & inhibitors , Cyclin D1/metabolism , DNA Methylation/drug effects , Enhancer Elements, Genetic , Histones/antagonists & inhibitors , Histones/genetics , Histones/metabolism , Humans , Lymphoma, Mantle-Cell/metabolism , Molecular Targeted Therapy , Transcription, Genetic/drug effects , Translocation, Genetic
10.
Ther Adv Hematol ; 3(5): 275-90, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23616915

ABSTRACT

Marginal zone lymphoma (MZL) is an indolent B-cell lymphoma arising from marginal zone B-cells present in lymph nodes and extranodal tissues. MZL comprises 5-17% of all non-Hodgkin's lymphomas in adults. The World Health Organization categorizes MZL into three distinct types based on their site of impact: (1) splenic marginal zone lymphoma (SMZL); (2) nodal marginal zone lymphoma (NMZL); (3) extranodal mucosa-associated lymphoid tissue (MALT) lymphoma, which can be subdivided into gastric and nongastric. The subgroups of MZL share some common features but are different in their biology and behavior. Owing to the rarity of MZL there are few randomized trials available comparing various treatment options and therefore treatment is controversial, lacking standard guidelines. Treatment should be patient tailored and can range from a 'watchful waiting' approach for asymptomatic patients without cytopenias to surgery or localized radiation therapy. Rituximab in combination with chemotherapy has resulted in longer failure-free survival than chemotherapy alone in patients with SMZL. Helicobacter pylori positive gastric MALT shows a good response rate to triple antibiotic therapy. Newer therapies such as bendamustine, everolimus, lenalidomide, vorinostat and phosphoinositide 3-kinase inhibitors are in clinical trials for patients with relapsed or refractory MZL and have shown promising results. We are presently conducting clinical trials testing the efficacy of the epigenetic activity of cladribine as a hypomethylating agent in combination with the histone deacetylase inhibitor (HDACi) vorinostat and rituximab in patients with MZL. Further studies with the newer agents should be done both in newly diagnosed or relapsed/refractory MZL to streamline the care and to avoid the use of toxic chemotherapies as initial treatment.

11.
Leuk Lymphoma ; 52(8): 1488-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21623691

ABSTRACT

Mantle cell lymphoma (MCL) is a non-Hodgkin lymphoma that is incurable with standard chemotherapy. There is no consensus on the best initial therapy, especially for elderly patients, who are not candidates for aggressive treatment approaches. Current National Comprehensive Cancer Network (NCCN) treatment guidelines include rituximab (R) plus cladribine for the initial treatment of MCL. However, few data are available to substantiate this recommendation. Therefore, to further define the role of R-cladribine for the initial treatment of MCL, we performed a retrospective chart review of 31 patients with MCL (median age, 67) treated with R-cladribine. The majority of responding patients also received R maintenance. The overall response rate was 87%, with 61% of patients achieving a complete remission (CR/CRu). The estimated median follow-up was 32.5 months, median PFS was 37.5 months, and median OS was 85.2 months. One of 19 (5.3%) subjects in CR/CRu relapsed (median follow-up of 23 months). CR/CRu was associated with improved survival (p < 0.0001), while a high mantle cell international prognostic index (MIPI) was associated with worse survival (p = 0.05). There was one toxic death (neutropenic pseudomonal sepsis) related to treatment. R-cladribine is an effective therapy for previously untreated MCL, and these results validate the use of R-cladribine for the initial treatment of MCL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, Mantle-Cell/drug therapy , Aged , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Cladribine/administration & dosage , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Lymphoma, Mantle-Cell/diagnosis , Male , Middle Aged , Prognosis , Remission Induction , Retrospective Studies , Rituximab , Time Factors , Treatment Outcome
12.
Clin Lymphoma Myeloma Leuk ; 10(6): 464-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21156463

ABSTRACT

BACKGROUND: Although the typical clinical manifestations of multiple myeloma (MM) are summarized by the CRAB symptoms (hypercalcemia, renal insufficiency, anemia, and bone lesions), a significant proportion of patients with MM present with a variety of other clinical manifestations. We conducted a study evaluating the presenting symptoms that led to the diagnosis of MM. PATIENTS AND METHODS: We conducted a retrospective review of 170 consecutive patients with MM seen at the Penn State Hershey Cancer Institute. RESULTS: Among patients with symptomatic MM, 74% presented with CRAB symptoms, 20% presented with non-CRAB manifestations, and 6% had both clinical features. Ten categories of non-CRAB manifestations were found, in order of decreasing frequency: neuropathy (because of spinal cord compression, nerve root compression, or peripheral neuropathy), extramedullary involvement, hyperviscosity syndrome, concomitant amyloidosis (eg, nephrotic syndrome or cardiopathy), hemorrhage/coagulopathy, systemic symptoms (eg, fever or weight loss), primary plasma cell leukemia, infections, cryoglobulinemia, and secondary gout. Kaplan-Meier estimates of survival in patients with non-CRAB manifestations did not show a significant difference from the survival of patients presenting with CRAB symptoms. CONCLUSION: Presenting symptoms of MM may be grouped in a total of 14 categories, 4 for the CRAB and 10 for the less common non-CRAB features. Grouped together, non-CRAB manifestations do not appear to confer a negative effect on the prognosis of patients with MM.


Subject(s)
Anemia/pathology , Bone and Bones/pathology , Hypercalcemia/pathology , Multiple Myeloma/diagnosis , Renal Insufficiency/pathology , Abnormalities, Multiple/classification , Abnormalities, Multiple/pathology , Adult , Aged , Aged, 80 and over , Diagnosis, Differential , Female , Femur , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Retrospective Studies , Syndrome
13.
Adv Hematol ; 2010: 601548, 2010.
Article in English | MEDLINE | ID: mdl-20672048

ABSTRACT

A single institution case series of adenovirus infections after allogeneic hematopoietic stem cell transplantation is presented to highlight the consideration for adenovirus infections as an etiology in patients with rapid hepatic or other sudden organ deterioration in the setting of apparent GVHD stabilization. The series also highlights that survival is limited with these infections often due in part to concomitant opportunistic infections. In addition, the pathophysiological events, such as GVHD and hepatic dysfunction, may complicate the clinical picture and delay therapy of an opportunistic infection. This is particularly true for adenoviral infections as they also have a distinct clinical picture in immunocompromised patients when compared to immune competent patients. Adenovirus infections also have the additional challenge that its treatment, cidofovir, has associated toxicities that can delay its administration. Recent developments has yielded an assay that can be used in the early detection and for serial determinations of adenovirus in patients with advanced GVHD, as well as a new therapeutic agent currently undergoing clinical trials.

14.
Expert Opin Investig Drugs ; 18(8): 1169-81, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19604118

ABSTRACT

Cladribine was synthesized as a purine analogue drug that inhibited adenosine deaminase. It received FDA approval in the 1980s for treatment of hairy cell leukemia. Given its toxicity towards lymphocytes and its corresponding immunosuppressive effects, it has been studied and found efficacious in a variety of hematologic malignancies and autoimmune conditions, most recently multiple sclerosis. This review highlights pharmacological, toxicological and clinical data for the use of cladribine. It also discusses existing and new mechanisms that may contribute to its unique clinical activity. Emerging data show that in addition to its known purine nucleoside analogue activity, cladribine possesses epigenetic properties, inhibiting S-adenosylhomocysteine hydrolase and DNA methylation. This may contribute to its efficacy and highlights the importance of studying combination therapy with other epigenetic or targeted agents. Clinical trials are underway in a variety of malignant and nonmalignant conditions.


Subject(s)
Adenosine Deaminase Inhibitors , Antineoplastic Agents/pharmacology , Cladribine/pharmacology , Purines/chemistry , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Autoimmune Diseases/drug therapy , Cladribine/adverse effects , Cladribine/chemistry , Cladribine/pharmacokinetics , Cladribine/therapeutic use , Clinical Trials as Topic , DNA Methylation/drug effects , Humans , Molecular Structure , Neoplasms/drug therapy
15.
J Exp Med ; 205(8): 1843-58, 2008 Aug 04.
Article in English | MEDLINE | ID: mdl-18625744

ABSTRACT

In mantle cell lymphoma (MCL) and some cases of multiple myeloma (MM), cyclin D1 expression is deregulated by chromosome translocations involving the immunoglobulin heavy chain (IgH) locus. To evaluate the mechanisms responsible, gene targeting was used to study long-distance gene regulation. Remarkably, these targeted cell lines lost the translocated chromosome (t(11;14)). In these MCL and MM cells, the nonrearranged cyclin D1 (CCND1) locus reverts from CpG hypomethylated to hypermethylated. Reintroduction of the translocated chromosome induced a loss of methylation at the unrearranged CCND1 locus, providing evidence of a transallelic regulatory effect. In these cell lines and primary MCL patient samples, the CCND1 loci are packaged in chromatin-containing CCCTC binding factor (CTCF) and nucleophosmin (NPM) at the nucleolus. We show that CTCF and NPM are bound at the IgH 3' regulatory elements only in the t(11;14) MCL cell lines. Furthermore, NPM short hairpin RNA produces a specific growth arrest in these cells. Our data demonstrate transvection in human cancer and suggest a functional role for CTCF and NPM.


Subject(s)
Genes, bcl-1 , Lymphoma, Mantle-Cell/genetics , Translocation, Genetic , CCCTC-Binding Factor , Cell Line, Tumor , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 14/genetics , CpG Islands , Cyclin D , Cyclins/genetics , Cyclins/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Gene Targeting , Genes, Immunoglobulin , Humans , Hybrid Cells , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Promoter Regions, Genetic , Repressor Proteins/metabolism
16.
Biol Blood Marrow Transplant ; 14(2): 246-55, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18215785

ABSTRACT

Allogeneic hematopoietic cell transplantation (HCT) is the only curative strategy for patients with myelodysplastic syndrome (MDS) and myeloproliferative disorders (MPD). We report the results of 148 patients (median age = 59 years old) with de novo MDS (n = 40), acute myelogenous leukemia (AML) after antecedent MDS/MPD (n = 49), treatment-related MDS (t-MDS) (n = 25), MPD (n = 27), and chronic myelomonocytic leukemia (CMML) (n = 7) who underwent allogeneic HCT using a conditioning regimen of low-dose total body irradiation (TBI) alone (200 cGy) on day 0 (n = 5) or with the addition of fludarabine (Flu) 30 mg/m(2)/day on days -4 to -2 (n = 143). Postgrafting immunosuppression consisted of cyclosporine and mycophenolate mofetil (MMF). Seventy-five patients (51%) received an allograft from a matched related donor (MRD), and 73 patients (49%) were recipients of unrelated donor (URD) grafts. There was no significant difference in the incidence of acute (gr II-IV) and chronic extensive graft-versus-host disease (aGVHD, cGVHD) between the recipients of related and unrelated donor grafts. By day +28, 75% of patients demonstrated mixed T cell chimerism. Graft rejection was seen in 15% of patients. With a median follow-up of 47 (range: 6-89) months, the 3-year relapse-free survival (RFS) and overall survival (OS) are both 27% for all patients, with a relapse incidence of 41%. The 3-year RFS for the patients with de novo MDS, AML after antecedent MDS/MPD, t-MDS, MPD, and CMML were 22%, 20%, 29%, 37%, and 43%, respectively, and the 3-year OS was 20%, 23%, 27%, 43%, and 43%, respectively. The 3-year nonrelapse mortality (NRM) was 32%. Factors associated with a lower risk of relapse were the development of extensive cGVHD and having a low risk or intermediate-1 risk International Prognostic Score for the de novo MDS patients. Nonmyeloablative HCT confers remissions in patients who otherwise were not eligible for conventional HCT but for whom relapse is the leading cause of treatment failure.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Myelodysplastic Syndromes/therapy , Myeloproliferative Disorders/therapy , Transplantation Conditioning/methods , Adult , Aged , Female , Humans , Leukemia, Myeloid, Acute/therapy , Leukemia, Myelomonocytic, Chronic/therapy , Male , Middle Aged , Survival Analysis , Transplantation, Homologous , Treatment Outcome , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Whole-Body Irradiation
17.
Blood ; 110(7): 2744-8, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17595333

ABSTRACT

Allogeneic hematopoietic cell transplantation (HCT) after nonmyeloablative conditioning for hematologic malignancies depends on graft-versus-tumor effects for eradication of cancer. Here, we estimated relapse risks according to disease characteristics. Between 1997 and 2006, 834 consecutive patients (median age, 55 years; range, 5-74 years) received related (n = 498) or unrelated (n = 336) HCT after 2 Gy total body irradiation alone (n = 171) or combined with fludarabine (90 mg/m(2); n = 663). Relapse rates per patient year (PY) at risk, corrected for follow-up and competing nonrelapse mortality, were calculated for 29 different diseases and stages. The overall relapse rate per PY was 0.36. Patients with chronic lymphocytic leukemia (CLL) and multiple myeloma (MM) in remission (CR), low-grade or mantle cell non-Hodgkin lymphoma (NHL) (CR + partial remission [PR]), and high-grade NHL-CR had the lowest rates (0.00-0.24; low risk). In contrast, patients with advanced myeloid and lymphoid malignancies had rates of more than 0.52 (high risk). Patients with lymphoproliferative diseases not in CR (except Hodgkin lymphoma and high-grade NHL) and myeloid malignancies in CR had rates of 0.26-0.37 (standard risk). In conclusion, patients with low-grade lymphoproliferative disorders experienced the lowest relapse rates, whereas patients with advanced myeloid and lymphoid malignancies had high relapse rates after nonmyeloablative HCT. The latter might benefit from cytoreductive treatment before HCT.


Subject(s)
Granulocyte Precursor Cells/pathology , Hematopoietic Stem Cell Transplantation , Leukemia/pathology , Leukemia/surgery , Transplantation Conditioning/methods , Adolescent , Adult , Aged , Cell Separation , Child , Child, Preschool , Female , Humans , Leukemia/classification , Leukemia/epidemiology , Male , Middle Aged , Recurrence , Risk , Survival Rate , Transplantation, Homologous
18.
Curr Treat Options Oncol ; 8(5): 375-81, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18214689

ABSTRACT

There is no consensus treatment for newly diagnosed mantle cell lymphoma. The CHOP + rituximab and hyperCVAD + rituximab regimens are most commonly used. The former is limited by relatively lower rates of complete remission (CR) and frequent relapses. The latter is limited by toxicities, especially in older patients, and relapses that occur later than those usually seen with CHOP + rituximab. Thus, improved therapies are needed. The purine analog cladribine (2-cda) + rituximab has been studied as an alternative frontline regimen in MCL and is quite active with minimal toxicity. Cladribine has epigenetic activity in that it inhibits DNA methylation. Cladribine + rituximab should be further studied in newly diagnosed mantle cell lymphoma in combination with new agents such as inhibitors of histone deacetylation, the mTOR pathway, and the proteasome.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Epigenesis, Genetic/drug effects , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/genetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cladribine/administration & dosage , Cyclophosphamide/administration & dosage , DNA Methylation , Dexamethasone/administration & dosage , Doxorubicin/administration & dosage , Humans , Lymphoma, Mantle-Cell/diagnosis , Prednisone/administration & dosage , Rituximab , Vincristine/administration & dosage
19.
J Clin Oncol ; 24(30): 4867-74, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-17001068

ABSTRACT

PURPOSE: Evaluate response rate, duration of response (DOR), time-to-progression (TTP), overall survival (OS), and safety of bortezomib treatment in patients with relapsed or refractory mantle cell lymphoma (MCL). PATIENTS AND METHODS: Bortezomib 1.3 mg/m(2) was administered on days 1, 4, 8, and 11 of a 21-day cycle, for up to 17 cycles. Response and progression were determined using International Workshop Response Criteria, both using data from independent radiology review and by the investigators. Primary efficacy analyses were based on data from independent radiology review. RESULTS: In total, 155 patients were treated. Median number of prior therapies was one (range, one to three). Response rate in 141 assessable patients was 33% including 8% complete response (CR)/unconfirmed CR. Median DOR was 9.2 months. Median TTP was 6.2 months. Results by investigator assessments were similar. Median OS has not been reached after a median follow-up of 13.4 months. The safety profile of bortezomib was similar to previous experience in relapsed multiple myeloma. The most common adverse events grade 3 or higher were peripheral neuropathy (13%), fatigue (12%), and thrombocytopenia (11%). Death from causes that were considered to be treatment related was reported for 3% of patients. CONCLUSION: These results confirm the activity of bortezomib in relapsed or refractory MCL, with predictable and manageable toxicities. Bortezomib provides significant clinical activity in terms of durable and complete responses, and may therefore represent a new treatment option for this population with usually very poor outcome. Studies of bortezomib-based combinations in MCL are ongoing.


Subject(s)
Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Lymphoma, Mantle-Cell/drug therapy , Pyrazines/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Boronic Acids/adverse effects , Bortezomib , Drug Resistance, Neoplasm , Female , Humans , Lymphoma, Mantle-Cell/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Pyrazines/adverse effects , Treatment Outcome
20.
PLoS Genet ; 2(2): e14, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16462943

ABSTRACT

Sox6 is a member of the Sox transcription factor family that is defined by the conserved high mobility group (HMG) DNA binding domain, first described in the testis determining gene, Sry. Previous studies have suggested that Sox6 plays a role in the development of the central nervous system, cartilage, and muscle. In the Sox6-deficient mouse, p100H, epsilony globin is persistently expressed, and increased numbers of nucleated red cells are present in the fetal circulation. Transfection assays in GM979 (erythroleukemic) cells define a 36-base pair region of the epsilony proximal promoter that is critical for Sox6 mediated repression. Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assays demonstrate that Sox6 acts as a repressor by directly binding to the epsilony promoter. The normal expression of Sox6 in wild-type fetal liver and the ectopic expression of epsilony in p100H homozygous fetal liver demonstrate that Sox6 functions in definitive erythropoiesis. The present study shows that Sox6 is required for silencing of epsilony globin in definitive erythropoiesis and suggests a role for Sox6 in erythroid cell maturation. Thus, Sox6 regulation of epsilony globin might provide a novel therapeutical target in the treatment of hemoglobinopathies such as sickle cell anemia and thalassemia.


Subject(s)
DNA-Binding Proteins/physiology , Erythropoiesis/genetics , Globins/genetics , High Mobility Group Proteins/physiology , Transcription Factors/physiology , Animals , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Erythrocytes/metabolism , Homozygote , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic , Protein Binding , SOXD Transcription Factors , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...