Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
NPJ Precis Oncol ; 6(1): 13, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35233056

ABSTRACT

The NCI-MATCH was designed to characterize the efficacy of targeted therapies in histology-agnostic driver mutation-positive malignancies. Sub-protocols F and G were developed to evaluate the role of crizotinib in rare tumors that harbored either ALK or ROS1 rearrangements. Patients with malignancies that progressed following at least one prior systemic therapy were accrued to the NCI-MATCH for molecular profiling, and those with actionable ALK or ROS1 rearrangements were offered participation in sub-protocols F or G, respectively. There were five patients who enrolled on Arm F (ALK) and four patients on Arm G (ROS1). Few grade 3 or 4 toxicities were noted, including liver test abnormalities, and acute kidney injury. For sub-protocol F (ALK), the response rate was 50% (90% CI 9.8-90.2%) with one complete response among the 4 eligible patients. The median PFS was 3.8 months, and median OS was 4.3 months. For sub-protocol G (ROS1) the response rate was 25% (90% CI 1.3-75.1%). The median PFS was 4.3 months, and median OS 6.2 months. Data from 3 commercial vendors showed that the prevalence of ALK and ROS1 rearrangements in histologies other than non-small cell lung cancer and lymphoma was rare (0.1% and 0.4% respectively). We observed responses to crizotinib which met the primary endpoint for ALK fusions, albeit in a small number of patients. Despite the limited accrual, some of the patients with these oncogenic fusions can respond to crizotinib which may have a therapeutic role in this setting.

2.
Obes Sci Pract ; 3(2): 212-218, 2017 06.
Article in English | MEDLINE | ID: mdl-28702214

ABSTRACT

INTRODUCTION: Co­existing decreased muscle mass and increased visceral fat, an age­associated change called sarcopenic obesity, results in fragility and cardiovascular disease. To assess the pathogenesis of sarcopenic obesity, we assessed the associations of clinical parameters with psoas muscle mass in elderly male subjects with obesity and type 2 diabetes. METHODS: The subjects were 55 patients, over 65 years of age and with a visceral fat area exceeding 100 cm2, with type 2 diabetes. The cross­sectional area of the psoas muscle is considered to provide an estimation of overall muscle mass. Sarcopenia was considered to be present when the total psoas muscle area was low, defined as a value below 500 mm2 m−2 on a computed tomographic scan. RESULTS: The maximum intima­media thickness (max IMT) and urinary 8­isoprostane values were significantly higher in the sarcopenic group. Multiple linear regression analysis revealed max IMT to be an independent variable related to muscle mass decline. In addition, logistic analysis showed max IMT and urinary 8­isoprostane to be variables independently contributing to total psoas muscle area <500 mm2 m−2. CONCLUSION: Worsening surrogate markers for systemic oxidative stress and atherosclerosis were associated with declining muscle mass in elderly subjects with obesity and type 2 diabetes. These results indicate that systemic oxidative stress is among the mechanisms underlying atherosclerosis development in subjects with sarcopenic obesity.

3.
Ann Oncol ; 27(2): 318-23, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26598548

ABSTRACT

BACKGROUND: Adenoid cystic carcinoma (ACC) is a subtype of malignant salivary gland tumors (MSGT), in which 90% of cases express cKIT. Dasatinib is a potent and selective inhibitor of five oncogenic protein tyrosine kinases (PTKs)/kinase families including cKIT. We conducted a phase II study to determine the antitumor activity of dasatinib in ACC and non-ACC MSGT. PATIENTS AND METHODS: In a two-stage design, patients with progressive, recurrent/metastatic ACC (+cKIT) and non-ACC MSGT (separate cohort) were treated with dasatinib 70 mg p.o. b.i.d. Response was assessed every 8 weeks using RECIST. RESULTS: Of 54 patients: 40 ACC, 14 non-ACC (1, ineligible excluded); M:F = 28 : 26, median age 56 years (range 20-82 years), ECOG performance status 0 : 1 : 2 = 24 : 28 : 2, prior radiation: 44, prior chemotherapy: 21. The most frequent adverse events (AEs) (as % of patients, worst grade 2 or higher) were: fatigue (28%), nausea (19%), headache (15%), lymphopenia (7%), dyspnea (11%), alanine aminotransferase increased (7%), anorexia (7%), vomiting (7%), alkaline phosphatase increased (6%), diarrhea (6%), neutropenia (6%), and noncardiac chest pain (6%). No grade 4 AE occurred, 15 patients experienced a grade 3 AE, primarily dyspnea (5) and fatigue (4), and cardiac toxicity (1 prolonged QTc). Among ACC patients, best response to dasatinib: 1 patient (2.5%) had partial response, 20 patients (50%) had stable disease (SD) (3-14 months), 12 patients (30%) had PD, 2 withdrew, 3 discontinued therapy due to AE, and 2 died before cycle 2. Median progression-free survival was 4.8 months. Median overall survival was 14.5 months. For 14 assessable non-ACC patients, none had objective response, triggering early stopping rule. Seven had SD (range 1-7 months), 4 PD, 2 discontinued therapy due to AE, and 1 died before cycle 2. CONCLUSION: Although there was only one objective response, dasatinib is well tolerated, with tumor stabilization achieved by 50% of ACC patients. Dasatinib demonstrated no activity in non-ACC MSGT.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Adenoid Cystic/drug therapy , Dasatinib/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Salivary Gland Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Carcinoma, Adenoid Cystic/pathology , Dasatinib/adverse effects , Disease-Free Survival , Female , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-kit/metabolism , Salivary Gland Neoplasms/pathology , Treatment Outcome , Young Adult
4.
Diabet Med ; 32(2): 213-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25255697

ABSTRACT

AIM: To analyse the effects of thyroid hormones on ß-cell function and glucose metabolism in people with prediabetes who are euthyroid. METHODS: A total of 111 people who were euthyroid underwent 75-g oral glucose tolerance tests, of whom 52 were assigned to the normal glucose tolerance and 59 to the prediabetes groups. Homeostatic model assessment of ß-cell function, insulinogenic index and areas under the curve for insulin and glucose were evaluated as indices of pancreatic ß-cell function. RESULTS: In both groups, BMI, fasting insulin, homeostasis model assessment ratio and HDL cholesterol correlated significantly with all indices of pancreatic ß-cell function. Free triiodothyronine correlated positively with all insulin secretion indices in the prediabetes group. Multiple linear regression analysis showed that free triiodothyronine was an independent variable that had a positive correlation with all indices of ß-cell function in the prediabetes group. By contrast, no such correlation was found in the normal glucose tolerance group. CONCLUSIONS: Free triiodothyronine is associated with both basal and glucose-stimulated insulin secretion in people with prediabetes who are euthyroid; therefore, the regulation of insulin secretion by thyroid hormones is a potentially novel therapeutic target for the treatment of diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Prediabetic State/physiopathology , Thyroid Gland/metabolism , Triiodothyronine/metabolism , Up-Regulation , Adult , Aged , Body Mass Index , Cholesterol, HDL/blood , Female , Glucose Tolerance Test , Humans , Insulin/blood , Insulin Resistance , Insulin Secretion , Male , Middle Aged , Overweight/complications , Prediabetic State/blood , Prediabetic State/complications , Prediabetic State/metabolism , Severity of Illness Index , Solubility , Triiodothyronine/blood , Triiodothyronine/chemistry
5.
Ann Oncol ; 24(11): 2922-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24170610

ABSTRACT

BACKGROUND: Pre-clinical data have suggested a therapeutic role of Hedgehog (Hh) pathway inhibitors in chondrosarcoma. METHODS: This phase II trial included patients with progressive advanced chondrosarcoma. They received GDC-0449 150 mg/day (days 1-28, 28-day cycle). The primary end point was the 6-month clinical benefit rate (CBR) defined as the proportion of patients with non-progressive disease at 6 months. A 6-month CBR of 40% was considered as a reasonable objective to claim drug efficacy. RESULTS: Between February 2011 and February 2012, 45 patients were included. Twenty had received prior chemotherapy. Thirty-nine were assessable for efficacy. The 6-month CBR was 25.6% (95% confidence interval 13.0-42.1). All stable patients had grade 1 or 2 conventional chondrosarcoma with documented progression within the 6 months before inclusion. All but one with available data also had overexpression of the Hh ligand. Median progression-free and overall survivals were 3.5 and 12.4 months, respectively. The most frequent adverse events were grade 1 or 2 myalgia, dysgeusia and alopecia. CONCLUSIONS: GDC-0449 did not meet the primary end point of this trial. Results suggest some activity in a subset of patients with progressive grade 1 or 2 conventional chondrosarcoma. Further studies assessing its role in combination with chemotherapy are warranted. CLINICALTRIALSGOV IDENTIFIER: NCT01267955.


Subject(s)
Anilides/administration & dosage , Chondrosarcoma/drug therapy , Drug-Related Side Effects and Adverse Reactions/pathology , Hedgehog Proteins/biosynthesis , Pyridines/administration & dosage , Adult , Aged , Aged, 80 and over , Chondrosarcoma/genetics , Chondrosarcoma/pathology , Disease-Free Survival , Drug-Related Side Effects and Adverse Reactions/classification , Female , France , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Humans , Male , Middle Aged
6.
Ann Oncol ; 23(12): 3151-3155, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22843420

ABSTRACT

BACKGROUND: Academic/institutional investigator-initiated clinical trials benefit individuals and society by supplementing gaps in industry-sponsored clinical trials. MATERIALS: In May 2010, experts from Japan, the Republic of Korea, the UK, and the United States, met at a symposium in Tokyo, Japan, to discuss how policies related to the conduct of clinical trials, which have been shown to be effective, may be applied to other regions of the world. RESULTS: In order to increase the availability of anticancer drugs world-wide, nations including Japan should examine the benefits of increasing the number of investigator-initiated clinical trials. These trials represent one of the most effective ways to translate basic scientific knowledge into clinical practice. These trials should be conducted under GCP guidelines and include Investigational New Drug application submissions with the ultimate goal of future drug approval. CONCLUSIONS: To maximize the effectiveness of these trials, a policy to educate health care professionals, cancer patients and their families, and the public in general on the benefits of clinical trials should be strengthened. Finally, policies that expedite the clinical development of novel cancer drugs which have already been shown to be effective in other countries are needed in many nations including Japan to accelerate drug approval.


Subject(s)
Clinical Trials as Topic/legislation & jurisprudence , Drug Discovery , Antineoplastic Agents , Drug Approval , Humans , Japan , Policy , Research Personnel
7.
Diabet Med ; 25(10): 1211-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19046200

ABSTRACT

AIMS: We prospectively studied Japanese workers with impaired fasting glucose (IFG) and/or impaired glucose tolerance (IGT) and analysed possible risk factors for diabetes, including psychosocial factors such as stress. METHODS: The participants were 128 male Japanese company employees (mean age, 49.3 +/- 5.9 years) with IFG and/or IGT diagnosed by oral glucose tolerance test (OGTT). Participants were prospectively studied for 5 years with annual OGTTs. The Kaplan-Meier method and Cox's proportional hazard model were used to analyse the incidence of diabetes and the factors affecting glucose tolerance, including anthropometric, biochemical and social-psychological factors. RESULTS: Of 128 participants, 36 (28.1%) developed diabetes and 39 (30.5%) returned to normal glucose tolerance (NGT) during a mean follow-up of 3.2 years. Independent risk factors for diabetes were night duty [hazard ratio (HR) = 5.48, P = 0.002], higher fasting plasma glucose (FPG) levels within 6.1-6.9 mmol/l (HR = 1.05, P = 0.031), stress (HR = 3.81, P = 0.037) and administrative position (HR = 12.70, P = 0.045), while independent factors associated with recovery were lower FPG levels (HR = 0.94, P = 0.017), being a white-collar worker (HR = 0.34, P = 0.033), non-smoking (HR = 0.31, P = 0.040) and lower serum alanine aminotransferase (ALT) levels (HR = 0.97, P = 0.042). CONCLUSIONS: In addition to FPG levels at baseline, psychosocial factors (night duty, stress and administrative position) are risk factors for Type 2 diabetes, while being a white-collar worker, a non-smoker and lower serum ALT levels are factors associated with return to NGT in Japanese workers with IFG and/or IGT.


Subject(s)
Diabetes Mellitus, Type 2/psychology , Glucose Intolerance/psychology , Prediabetic State/psychology , Adult , Chi-Square Distribution , Disease Progression , Humans , Life Style , Male , Middle Aged , Multivariate Analysis , Prospective Studies , Psychology , Risk Factors , Smoking , Stress, Psychological
8.
Bone Marrow Transplant ; 37(1): 65-72, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16247422

ABSTRACT

Although high-dose therapy and autologous stem cell transplant (ASCT) is superior to conventional chemotherapy for treatment of myeloma, most patients relapse and the time to relapse depends upon the initial prognostic factors. The administration of non-cross-resistant chemotherapies during the post-transplant period may delay or prevent relapse. We prospectively studied the role of consolidation chemotherapy (CC) after single autologous peripheral blood stem cell transplant (auto-PBSCT) in 103 mostly newly diagnosed myeloma patients (67 patients were < or =6 months from the initial treatment). Patients received conditioning with BCNU, melphalan+/-gemcitabine and auto-PBSCT followed by two cycles of the DCEP+/-G regimen (dexamethasone, cyclophosphamide, etoposide, cisplatin+/-gemcitabine) at 3 and 9 months post-transplant and alternating with two cycles of DPP regimen (dexamethasone, cisplatin, paclitaxel) at 6 and 12 months post-transplant. With a median follow-up of 61.2 months, the median event-free survival (EFS) and overall survival (OS) are 26 and 54.1 months, respectively. The 5-year EFS and OS are 23.1 and 42.5%, respectively. Overall, 51 (49.5%) patients finished all CC, suggesting that a major limitation of this approach is an inability to deliver all planned treatments. In order to improve results following autotransplantation, novel agents or immunologic approaches should be studied in the post-transplant setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Multiple Myeloma/therapy , Peripheral Blood Stem Cell Transplantation , Transplantation Conditioning , Adult , Aged , Cisplatin , Combined Modality Therapy/methods , Cyclophosphamide , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dexamethasone , Disease-Free Survival , Etoposide , Female , Follow-Up Studies , Humans , Immunosuppressive Agents/administration & dosage , Male , Melphalan/administration & dosage , Middle Aged , Multiple Myeloma/mortality , Myeloablative Agonists/administration & dosage , Prospective Studies , Transplantation, Autologous , Gemcitabine
9.
Bone Marrow Transplant ; 34(10): 883-90, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15517008

ABSTRACT

Relapse remains a major cause of treatment failure after autotransplantation (auto-PBSCT) for Hodgkin's disease (HD). The administration of non-crossresistant therapies during the post-transplant period may delay or prevent relapse. We prospectively studied the role of consolidation chemotherapy (CC) after auto-PBSCT in 37 patients with relapsed or refractory HD. Patients received high-dose gemcitabine-BCNU-melphalan and auto-PBSCT followed by involved-field radiation and up to four cycles of the DCEP-G regimen, which consisted of dexamethasone, cyclophosphamide, etoposide, cisplatin, gemcitabine given at 3 and 9 months post transplant alternating with a second regimen (DPP) of dexamethasone, cisplatin, paclitaxel at 6 and 12 months post transplant. The probabilities of event-free survival (EFS) and overall survival (OS) at 2.5 years were 59% (95% CI=42-76%) and 86% (95% CI=71-99%), respectively. In all, 17 patients received 54 courses of CC and 15 were surviving event free (2.5 years, EFS=87%). There were no treatment-related deaths during or after the CC phase. Post-transplant CC is feasible and well tolerated. The impact of this approach on EFS should be evaluated in a larger, randomized study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Hodgkin Disease/therapy , Salvage Therapy/methods , Adolescent , Adult , Cisplatin/administration & dosage , Cyclophosphamide/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dexamethasone/administration & dosage , Etoposide/administration & dosage , Female , Hematopoietic Stem Cell Transplantation/mortality , Hodgkin Disease/mortality , Humans , Male , Middle Aged , Salvage Therapy/mortality , Secondary Prevention , Survival Analysis , Transplantation, Autologous , Gemcitabine
10.
Biol Blood Marrow Transplant ; 10(7): 473-83, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15205668

ABSTRACT

This study was conducted to define a new maximum tolerated dose and the dose-limiting toxicity (DLT) of melphalan and autologous hematopoietic stem cell transplantation (AHSCT) when used with the cytoprotective agent amifostine. Fifty-eight patients with various types of malignancy who were ineligible for higher-priority AHSCT protocols were entered on a phase I study of escalating doses of melphalan beginning at 220 mg/m(2) and advancing by 20 mg/m(2) increments in planned cohorts of 4 to 8 patients until severe regimen-related toxicity (RRT) was encountered. In all patients, amifostine 740 mg/m(2) was given on 2 occasions before the first melphalan dose (ie, 24 hours before and again 15 minutes before). AHSCT was given 24 hours after the first melphalan dose. Melphalan was given in doses up to and including 300 mg/m(2). Hematologic depression was profound, although it was rapidly and equally reversible at all melphalan doses. Although mucosal RRT was substantial, it was not the DLT, and some patients given the highest melphalan doses (ie, 300 mg/m(2)) did not develop mucosal RRT. The DLT was not clearly defined. Cardiac toxicity in the form of atrial fibrillation occurred in 3 of 36 patients treated with melphalan doses >/=280 mg/m(2) and was deemed fatal in 1 patient given melphalan 300 mg/m(2). (Another patient with a known cardiomyopathy was given melphalan 220 mg/m(2) and died as a result of heart failure but did not have atrial fibrillation.) Another patient given melphalan 300 mg/m(2) died of hepatic necrosis. The maximum tolerated dose of melphalan in this setting was thus considered to be 280 mg/m(2), and 27 patients were given this dose without severe RRT. Moreover, 38 patients were evaluable for delayed toxicity related to RRT; none was noted. Tumor responses have been noted at all melphalan doses and in all diagnostic groups, and 21 patients are alive at median day +1121 (range, day +136 to day +1923), including 16 without evidence of disease progression at median day +1075 (range, day +509 to day +1638). Amifostine and AHSCT permit the safe use of melphalan 280 mg/m(2), an apparent increase over the dose of melphalan that can be safely administered with AHSCT but without amifostine. Further studies are needed not only to confirm these findings, but also to define the antitumor efficacy of this regimen. Finally, it may be possible to evaluate additional methods of further dose escalation of melphalan in this setting.


Subject(s)
Amifostine/administration & dosage , Drug-Related Side Effects and Adverse Reactions , Hematopoietic Stem Cell Transplantation , Maximum Tolerated Dose , Melphalan/administration & dosage , Neoplasms/therapy , Adult , Aged , Cohort Studies , Disease-Free Survival , Female , Humans , Male , Melphalan/adverse effects , Middle Aged , Transplantation, Autologous , Treatment Outcome
11.
Bone Marrow Transplant ; 34(1): 69-76, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15133484

ABSTRACT

The purpose of the study was to examine the yield of CD34(+) cells, response rates, and toxicity of high-dose cyclophosphamide with or without etoposide in patients with multiple myeloma. In total, 77 myeloma patients received either cyclophosphamide 4.5 g/m(2) (n=28) alone or with etoposide 2 g/m(2) (n=49) in a nonrandomized manner, followed by G-CSF 10 microg/kg/day for the purpose of stem cell mobilization. The effects of various factors on CD34(+) cell yield, response rate and engraftment were explored. A median of 22.39 x 10(6) CD34(+) cells/kg were collected on the first day of leukapheresis (range 0.59-114.71 x 10(6)/kg) in 71 (92%) of patients. Greater marrow plasma cell infiltration (P=0.02) or prior radiation therapy (P=0.02) adversely affected CD34(+) cell yield. In total, 45% of patients receiving cyclophosphamide and 56% of those receiving cyclophosphamide/etoposide had at least a minimum response by EBMT criteria. In all, 25% of patients who received cyclophosphamide alone vs 75.5% of patients who received combined chemotherapy required hospitalization mainly for treatment of neutropenic fever. Cyclophosphamide alone is associated with impressive CD34(+) cell yields and clear antimyeloma activity. The addition of etoposide resulted in increased toxicity without significant improvement in CD34(+) cell yield or response rates.


Subject(s)
Cyclophosphamide/administration & dosage , Etoposide/administration & dosage , Hematopoietic Stem Cell Mobilization/methods , Multiple Myeloma/therapy , Peripheral Blood Stem Cell Transplantation/methods , Adult , Aged , Antigens, CD34/analysis , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/toxicity , Cyclophosphamide/toxicity , Etoposide/toxicity , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization/standards , Humans , Leukapheresis/methods , Male , Middle Aged , Multiple Myeloma/complications , Peripheral Blood Stem Cell Transplantation/adverse effects , Treatment Outcome
12.
Bone Marrow Transplant ; 33(8): 781-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14767498

ABSTRACT

High-dose chemotherapy using melphalan (HDMEL) is an important component of many conditioning regimens that are given before autologous hematopoietic stem cell transplantation (AHSCT). In contrast to the situation in myeloma, and to a lesser degree acute leukemia, only a very limited published experience exists with the use of HDMEL conditioning as a single agent in doses requiring AHSCT for lymphoma, both Hodgkin lymphoma (HL) and especially non-Hodgkin lymphoma (NHL). Thus, we report results of treating 26 lymphoma patients (22 with NHL and four with HL) with HDMEL 220-300 mg/m(2) plus amifostine (AF) cytoprotection and AHSCT as part of a phase I-II trial. Median age was 51 years (range 24-62 years); NHL histology was varied, but was aggressive (including transformed from indolent) in 19 patients, indolent in two patients and mantle cell in one. All 26 patients had been extensively treated; 11 were refractory to the immediate prior therapy on protocol entry and two had undergone prior AHSCT. All were deemed ineligible for other, 'first-line' AHSCT regimens. Of these 26 patients, 22 survived to initial tumor evaluation on D +100. At this time, 13 were in complete remission, including four patients who were in second CR before HDMEL+AF+AHSCT. Responses occurred at all HDMEL doses. Currently, seven patients are alive, including five without progression, with a median follow-up in these latter patients of D +1163 (range D +824 to D +1630); one of these patients had a nonmyeloablative allograft as consolidation on D +106. Conversely, 14 patients relapsed or progressed, including five who had previously achieved CR with the AHSCT procedure. Two patients, both with HL, remain alive after progression; one is in CR following salvage radiotherapy. Six patients died due to nonrelapse causes, including two NHL patients who died while in CR. We conclude that HDMEL+AF+AHSCT has significant single-agent activity in relapsed or refractory NHL and HL. This experience may be used as a starting point for subsequent dose escalation of HDMEL (probably with AF) in established combination regimens.


Subject(s)
Amifostine/administration & dosage , Antineoplastic Agents, Alkylating/administration & dosage , Hematopoietic Stem Cell Transplantation , Hodgkin Disease/therapy , Lymphoma, Non-Hodgkin/therapy , Melphalan/administration & dosage , Radiation-Protective Agents/administration & dosage , Adult , Combined Modality Therapy , Female , Humans , Male , Middle Aged , Transplantation Conditioning/methods , Transplantation, Autologous
13.
Bone Marrow Transplant ; 29(4): 303-12, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11896427

ABSTRACT

Disease relapse occurs in 50% or more of patients who are autografted for relapsed or refractory lymphoma (NHL) or Hodgkin's disease (HD). The administration of non-cross-resistant therapies during the post-transplant phase could possibly control residual disease and delay or prevent its progression. To test this approach, 55 patients with relapsed/refractory or high-risk NHL or relapsed/refractory HD were enrolled in the following protocol: stem cell mobilization: cyclophosphamide (4.5 g/m(2)) + etoposide (2.0 g/m(2)) followed by GM-CSF or G-CSF; high-dose therapy: gemcitabine (1.0 g/m(2)) on day -5, BCNU (300 mg/m(2)) + gemcitabine (1.0 g/m(2)) on day -2, melphalan (140 mg/m(2)) on day -1, blood stem cell infusion on day 0; post-transplant immunotherapy (B cell NHL): rituxan (375 mg/m(2)) weekly for 4 weeks + GM-CSF (250 microg thrice weekly) (weeks 4-8); post-transplant involved-field radiotherapy (HD): 30-40 Gy to pre-transplant areas of disease (weeks 4-8); post-transplant consolidation chemotherapy (all patients): dexamethasone (40 mg daily)/cyclophosphamide (300 mg/m(2)/day)/etoposide (30 mg/m(2)/day)/cisplatin (15 mg/m(2)/day) by continuous intravenous infusion for 4 days + gemcitabine (1.0 g/m(2), day 3) (months 3 + 9) alternating with dexamethasone/paclitaxel (135 mg/m(2))/cisplatin (75 mg/m(2)) (months 6 + 12). Of the 33 patients with B cell lymphoma, 14 had primary refractory disease (42%), 12 had relapsed disease (36%) and seven had high-risk disease in first CR (21%). For the entire group, the 2-year Kaplan-Meier event-free survival (EFS) and overall survival (OS) were 30% and 35%, respectively, while six of 33 patients (18%) died before day 100 from transplant-related complications. The rituxan/GM-CSF phase was well-tolerated by the 26 patients who were treated and led to radiographic responses in seven patients; an eighth patient with a blastic variant of mantle-cell lymphoma had clearance of marrow involvement after rituxan/GM-CSF. Of the 22 patients with relapsed/refractory HD (21 patients) or high-risk T cell lymphoblastic lymphoma (one patient), the 2-year Kaplan-Meier EFS and OS were 70% and 85%, respectively, while two of 22 patients (9%) died before day 100 from transplant-related complications. Eight patients received involved field radiation and seven had radiographic responses within the treatment fields. A total of 72 courses of post-transplant consolidation chemotherapy were administered to 26 of the 55 total patients. Transient grade 3-4 myelosuppression was common and one patient died from neutropenic sepsis, but no patients required an infusion of backup stem cells. After adjustment for known prognostic factors, the EFS for the cohort of HD patients was significantly better than the EFS for an historical cohort of HD patients autografted after BEAC (BCNU/etoposide/cytarabine/cyclophosphamide) without consolidation chemotherapy (P = 0.015). In conclusion, post-transplant consolidation therapy is feasible and well-tolerated for patients autografted for aggressive NHL and HD and may be associated with improved progression-free survival particularly for patients with HD.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hodgkin Disease/therapy , Lymphoma, Non-Hodgkin/therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Combined Chemotherapy Protocols , Combined Modality Therapy , Disease-Free Survival , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Hodgkin Disease/drug therapy , Hodgkin Disease/pathology , Hodgkin Disease/radiotherapy , Humans , Immunotherapy , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/pathology , Lymphoma, Non-Hodgkin/radiotherapy , Male , Middle Aged , Rituximab , Transplantation, Autologous
14.
Mol Ther ; 3(1): 88-96, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11162315

ABSTRACT

The genetic transfer of drug resistance to hematopoietic cells is an attractive approach to overcoming myelosuppression caused by high-dose chemotherapy. Because cyclophosphamide (CTX) and methotrexate (MTX) are commonly used non-cross-resistant drugs, generation of dual drug resistance in hematopoietic cells that allows dose intensification may increase anti-tumor effects and circumvent the emergence of drug-resistant tumors. We constructed a retroviral vector containing both a human cytosolic ALDH-1 cDNA and a human doubly mutated DHFR cDNA (Phe22/Ser31; termed F/S in the description of constructs) to generate increased resistance to both CTX and MTX. Infection of NIH3T3 cells resulted in increased resistance to both 4-hydroperoxycyclophosphamide (4HC) (1.9 +/- 0.1-fold) and MTX (73 +/- 2.8-fold). Transduced human CD34(+) enriched hematopoietic progenitor cells were also resistant to both 4HC and MTX by CFU-GM readout. Lethally irradiated mice transplanted with SFG-ALDH-IRES-F/S or mock-transduced bone marrow cells were treated with high-dose pulse CTX or high-dose CTX/MTX. Animals receiving marrow not transduced with ALDH-1 or mutated DHFR cDNA died from CTX or CTX/MTX toxicity, whereas mice transduced with ALDH-1 and mutated DHFR cDNA-containing marrow were able to tolerate the same doses of CTX or CTX/MTX treatment posttransplant. These data taken together indicate that ALDH-1 overexpression and mutant DHFR increased both 4HC and MTX resistance in vitro and in the in vivo mouse model. This construct may be useful for protecting patients from high-dose CTX- and MTX-induced myelosuppression.


Subject(s)
Aldehyde Dehydrogenase/genetics , Antimetabolites, Antineoplastic/pharmacology , Cyclophosphamide/pharmacology , Drug Resistance, Neoplasm/genetics , Methotrexate/pharmacology , Mutation , Retroviridae/genetics , Tetrahydrofolate Dehydrogenase/genetics , 3T3 Cells , Animals , Antigens, CD34/metabolism , Blotting, Western , Bone Marrow Transplantation , Cyclophosphamide/analogs & derivatives , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Gene Transfer Techniques , Hematopoietic Stem Cells/metabolism , Humans , Mice , Models, Genetic , Polymerase Chain Reaction , RNA/metabolism , Time Factors , Transduction, Genetic
15.
Cancer Gene Ther ; 8(12): 966-73, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11781659

ABSTRACT

Severe 5-fluorouracil (5-FU) toxicity has been reported among patients lacking dihydropyrimidine dehydrogenase (DPD) enzymatic activity. DPD is the principal enzyme involved in the degradation of 5-FU to 5'-6'-dihydrofluorouracil, which is further metabolized to fluoro-beta-alanine. We demonstrate here that overexpression of human DPD confers resistance to 5-FU in NIH3T3 cells, mouse bone marrow cells, and in human CD34+-enriched hematopoietic progenitor cells. An SFG-based dicistronic retroviral vector containing human DPD cDNA, an internal ribosomal entry site (IRES), and the neomycin phosphotransferase (Neo) gene was constructed (SFG-DPD-IRES-Neo). Transduced NIH3T3 cells demonstrated a 2-fold (ED50) increase in resistance to a 4-hour exposure of 5-FU in comparison to nontransduced cells. Expression of DPD was confirmed by Northern and Western blot analyses, and DPD enzyme activity was detectable only in transduced cells. Infection of mouse bone marrow cells with this retroviral construct resulted in an increased number of 5-FU-resistant CFU-GM colonies, compared to mock-transduced bone marrow in both 4-hour and 12- to 14-day exposures. Infection of human CD34+-enriched cells with this construct and incubation with 5-FU (10(-6) M) for 14 days also resulted in an increased number of 5-FU-resistant colonies. Retroviral transduction of human hematopoietic progenitor cells with a cDNA-expressing human DPD conferred resistance to 5-FU in NIH3T3 cells, mouse bone marrow cells, and human CD34+-enriched cells. These results encourage the use of this gene as a method to protect patients from 5-FU myelotoxicity.


Subject(s)
Drug Resistance/genetics , Hematopoietic Stem Cells/physiology , Oxidoreductases/genetics , 3T3 Cells , Animals , Antimetabolites, Antineoplastic/toxicity , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Dihydrouracil Dehydrogenase (NADP) , Fluorouracil/toxicity , Genetic Vectors , Hematopoietic Stem Cells/drug effects , Humans , Immunosuppressive Agents/toxicity , Mice , Oxidoreductases/biosynthesis , Retroviridae , Transfection
16.
Cancer Gene Ther ; 7(6): 910-9, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10880023

ABSTRACT

We previously reported the protection of hematopoietic cells from methotrexate (MTX) toxicity using an N2-based double copy vector containing serine 31 (S31)-mutated dihydrofolate reductase (DHFR) (DC/SV6S31). To examine whether the use of SFG-based dicistronic vectors will lead to improvement in gene transfer over the DC/SV6 vector, we compared the protection provided by MTX to NIH3T3 cells and hematopoietic progenitor cells infected with these retroviral constructs containing the S31 variant DHFR cDNA. In NIH3T3 cells, the 50% effective dose values of MTX conferred by the SFG vector were 8-fold higher than those obtained with the DC/SV6 vector. DHFR mRNA levels were 22-fold and 38-fold higher than that seen for the DC/SV6 vector according to Northern blot and real-time polymerase chain reaction analysis, respectively. However, DHFR protein expression and DHFR enzyme activity were only 1.5-fold and 2-fold higher in the SFG vector, respectively, indicating that the mRNA from the SFG vector is translated less efficiently than the mRNA generated from the DC/SV6 vector. Furthermore, the degree of MTX protection conferred by each vector in both mouse and human hematopoietic cells was the same. These results indicate that the in vitro transduction efficiency and transgene expression of human DHFR in hematopoietic progenitor cells is equally conferred by both vectors.


Subject(s)
3T3 Cells/drug effects , Bone Marrow/metabolism , DNA, Complementary/genetics , Drug Resistance/genetics , Hematopoietic Stem Cells/metabolism , Methotrexate/pharmacology , Moloney murine leukemia virus/genetics , Tetrahydrofolate Dehydrogenase/genetics , Animals , Blotting, Northern , Blotting, Southern , Blotting, Western , Bone Marrow/drug effects , DNA Primers/chemistry , Gene Expression Regulation, Enzymologic , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Hematopoietic Stem Cells/drug effects , Humans , Leukemia/pathology , Male , Mice , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Tetrahydrofolate Dehydrogenase/metabolism , Transduction, Genetic , Tumor Cells, Cultured
17.
Gynecol Oncol ; 76(3): 305-10, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10684701

ABSTRACT

We investigated whether retrovirus-mediated transfer of the herpes simplex thymidine kinase gene into a human endometrial carcinoma (EC4) cell line can sensitize these cells to the prodrug ganciclovir (GCV) and thereby provide a therapeutic option for this cancer. A retrovirus encoding for the herpes simplex virus tip-1 (HSV) thymidine kinase (tk) gene was generated in which expression of tk is under control of the myeloproliferative sarcoma virus (MPSV) promoter/enhancer. We used human mutated dihydrofolate reductase (DHFR) cDNA as a selectable marker. Expression of tk was confirmed by Northern blot analysis and reverse transcription polymerase chain reaction. We demonstrated that the combination of retrovirally mediated tk gene transfer and GCV treatment effectively inhibits proliferation and causes death of EC4 cells in vitro. A bystander killing effect was observed when 90% of uninfected tumor cells were mixed with only 10% of HSVtk-infected cells. We suggest that a gene therapy approach to endometrial carcinoma can be established using retroviral transfer of HSVtk to tumor cells and subsequent administration of GCV.


Subject(s)
Antiviral Agents/therapeutic use , Endometrial Neoplasms/therapy , Ganciclovir/therapeutic use , Genes, Viral , Genetic Therapy , Herpes Simplex/genetics , Prodrugs/therapeutic use , Thymidine Kinase/genetics , Drug Screening Assays, Antitumor , Female , Gene Transfer Techniques , Genetic Vectors , Humans , RNA, Messenger/analysis , Retroviridae , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
19.
J Interferon Cytokine Res ; 18(5): 321-6, 1998 May.
Article in English | MEDLINE | ID: mdl-9620359

ABSTRACT

Interleukin-1 (IL-1) produced in peripheral blood mononuclear cell (PBMC) cultures or added exogenously has been shown to upregulate HIV expression in vitro. Inhibition of IL-1 in HIV-infected individuals may inhibit HIV activation and slow disease progression. Recombinant human IL-1 receptor (rHu-IL-1R), the soluble extracellular portion of the human type I IL-1 receptor, inhibits HIV expression in acutely infected primary PBMCs and in the chronically infected promonocytic cell line, U1. We, therefore, conducted a phase I/II trial of the soluble rHu-IL-1R in HIV-1-infected individuals with CD4 T cell counts <300/microl to evaluate its safety and activity. Twelve evaluable patients were enrolled at three rHu-IL-1R dose levels:125 (n=3), 500 (n=3), and 1250 (n=6) microg/m2 per dose by subcutaneous (s.c.) injection three times a week for 8 weeks, followed by a 4 week observation period. rHu-IL-1R was safe and well tolerated. There were no deaths, no treatment-related grade 3/4 events, and no premature study discontinuations because of adverse events. The maximum tolerated dose was not reached. Seven patients reported improvements in one or more symptoms, including weight gain (3), improved energy level (4), decreased diarrhea (1), decreased night sweats (1), improvement in psoriatic arthritis (1), and improvement in a nonspecific chronic diffuse skin rash (1). Of 3 evaluable patients with Kaposi's sarcoma, 1 remained stable and 2 showed minimal progression. No consistent trends in absolute CD4 counts or percentages, quantitative HIV cultures, or serum p24 antigen, beta2-microglobulin, or triglyceride levels were observed. rHu-IL-1R is safe and well tolerated at the doses tested but induced no consistent changes in objective markers of HIV disease. Symptomatic improvements will require confirmation in randomized, placebo-controlled trials.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , HIV-1 , Receptors, Interleukin-1/therapeutic use , Adult , CD4 Lymphocyte Count , Cells, Cultured , Female , Humans , Male , Middle Aged , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Solubility
20.
Cancer Gene Ther ; 5(3): 144-9, 1998.
Article in English | MEDLINE | ID: mdl-9622097

ABSTRACT

We have previously shown that successful gene transfer of a mutated dihydrofolate reductase (DHFR) cDNA confers resistance to methotrexate (MTX) upon infected cells. We constructed a retrovirus vector, DC/SV6S31GPT, which carries both the Escherichia coli xanthine-guanine phosphoribosyltransferase gene and the mutated Serine 31 DHFR gene. Mouse fibroblast NIH3T3 cells infected with DC/SV6S31 GPT are more resistant to MTX than cells infected with DC/SV6S31, which carries the Serine 31 DHFR and the neomycin resistance gene cDNA. The mechanism of this augmented resistance is the increased salvaging of purines due to expression of xanthine-guanine phosphoribosyltransferase, as the augmentation does not occur when dialyzed serum, containing little xanthine or guanine, is used for cytotoxicity assays. These results indicate that coexpression of a metabolically related gene can potentiate the resistance carried by a drug resistance gene. This vector may be useful in clinical gene therapy to protect bone marrow from the toxic effects of MTX.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Gene Transfer Techniques , Methotrexate/pharmacology , Pentosyltransferases/genetics , Purines/metabolism , Tetrahydrofolate Dehydrogenase/genetics , 3T3 Cells , Animals , Base Sequence , Blotting, Northern , Bone Marrow Cells , DNA Primers , Drug Resistance, Neoplasm , Genetic Vectors , Male , Mice , Mice, Inbred C57BL , Retroviridae/genetics , Thioguanine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...