Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 742
Filter
Add more filters

Publication year range
1.
Br J Haematol ; 195(4): 571-584, 2021 11.
Article in English | MEDLINE | ID: mdl-34405395

ABSTRACT

Adult T-cell leukaemia/lymphoma (ATL) patients have a poor prognosis. Here, we investigated the impact of TP53 gene mutations on prognosis of ATL treated in different ways. Among 177 patients, we identified 47 single nucleotide variants or insertion-deletions (SNVs/indels) of the TP53 gene in 37 individuals. TP53 copy number variations (CNVs) were observed in 38 patients. Altogether, 67 of 177 patients harboured TP53 SNVs/indels or TP53 CNVs, and were categorized as having TP53 mutations. In the entire cohort, median survival of patients with and without TP53 mutations was 1·0 and 6·7 years respectively (P < 0·001). After allogeneic haematopoietic stem cell transplantation (HSCT), median survival of patients with (n = 16) and without (n = 29) TP53 mutations was 0·4 years and not reached respectively (P = 0·001). For patients receiving mogamulizumab without allogeneic HSCT, the median survival from the first dose of antibody in patients with TP53 mutations (n = 27) was only 0·9 years, but 5·1 years in those without (n = 42; P < 0·001). Thus, TP53 mutations are associated with unfavourable prognosis of ATL, regardless of treatment strategy. The establishment of alternative modalities to overcome the adverse impact of TP53 mutations in patients with ATL is required.


Subject(s)
Genes, p53 , Leukemia-Lymphoma, Adult T-Cell/genetics , Mutation , Adult , Aged , Aged, 80 and over , Allografts , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD28 Antigens/genetics , Carboplatin/administration & dosage , Cyclophosphamide/administration & dosage , DNA Copy Number Variations , Doxorubicin/administration & dosage , Etoposide/administration & dosage , Female , Hematopoietic Stem Cell Transplantation , Humans , INDEL Mutation , Kaplan-Meier Estimate , Lenalidomide/administration & dosage , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/mortality , Leukemia-Lymphoma, Adult T-Cell/therapy , Male , Middle Aged , Nitrosourea Compounds/administration & dosage , Polymorphism, Single Nucleotide , Prednisolone/administration & dosage , Prednisone/administration & dosage , Prognosis , Receptors, CCR4/genetics , Vincristine/administration & dosage , Vindesine/administration & dosage
2.
Cancer Sci ; 111(12): 4567-4580, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32976684

ABSTRACT

Adult T-cell leukemia/lymphoma (ATL) is a mature T-cell neoplasm and is classified into four subtypes (acute, lymphoma, chronic, and smoldering) according to the Shimoyama classification, established in 1991 through several nationwide surveys based on the clinical diversity of patients diagnosed in 1983-1987 in Japan. Thereafter, no such studies have been conducted. Recently, we conducted a nationwide hospital survey using the method of the 1980s studies, collected baseline data on 996 ATL patients diagnosed in 2010-2011 from 126 hospitals, and reported their unique epidemiological characteristics. Here, we report the follow-up results of registered ATL patients with the goal of evaluating current prognoses and treatment modalities as of 2016-2017. Of 770 evaluable patients, 391 (50.8%) had acute-type, 192 (24.9%) had lymphoma-type, 106 (13.8%) had chronic-type, and 81 (10.5%) had smoldering-type ATL. The initial therapy regimens used for acute/lymphoma-type ATL were vincristine, cyclophosphamide, doxorubicin and prednisone, followed by doxorubicin, ranimustine, and prednisone and then by vindesine, etoposide, carboplatin, and prednisone (VCAP-AMP-VECP)-like in 38.5/41.7% and cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP)-like in 14.6/13.7% of patients. Allogeneic hematopoietic stem cell transplantation was used to treat 15.9/10.4% of acute/lymphoma-type ATL patients. The 4-year survival rates (the median survival time, days) for acute-, lymphoma-, unfavorable chronic-, favorable chronic-, and smoldering-type ATL were 16.8% (252), 19.6% (305), 26.6% (572), 62.1% (1937), and 59.8% (1851), respectively. The 4-year survival rates for acute- and lymphoma-type ATL improved compared with those reported in 1991, but those for chronic- and smoldering-type ATL were not. Further efforts are warranted to develop more efficient therapeutic strategies to improve the prognosis of ATL in Japan.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation , Leukemia-Lymphoma, Adult T-Cell/mortality , Leukemia-Lymphoma, Adult T-Cell/therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cause of Death , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Health Care Surveys/statistics & numerical data , Hospitals/statistics & numerical data , Humans , Japan/epidemiology , Leukemia-Lymphoma, Adult T-Cell/blood , Leukemia-Lymphoma, Adult T-Cell/classification , Male , Middle Aged , Nitrosourea Compounds/administration & dosage , Prednisone/administration & dosage , Prognosis , Retrospective Studies , Survival Rate , Vincristine/administration & dosage , Vindesine/administration & dosage
3.
BMC Cancer ; 18(1): 552, 2018 May 10.
Article in English | MEDLINE | ID: mdl-29747595

ABSTRACT

BACKGROUND: It is frequently asked whether chemotherapy can still play a role in metastatic melanoma considering the effectiveness of the available drugs today, including antiCTLA4/antiPD1 immunotherapy and antiBRAF/antiMEK inhibitors. However, only approximately half of patients respond to these drugs, and the majority progress after 6-11 months. Therefore, a need for other therapeutic options is still very much apparent. We report the first large trial of a sequential full dose of fotemustine (FM) preceded by a low dose of temozolomide (TMZ) as a chemo-modulator in order to inactivate the DNA repair action of O(6)-methylguanine DNA-methyltransferase (MGMT). Primary endpoints were overall response and safety. We also evaluated specific biological parameters aiming to tailor these chemotherapies to selected patients. METHODS: A total of 69 consecutive patients were enrolled. The main features included a median age of 60 years (21-81) and M1c stage, observed in 74% of the patients, with brain metastases in 15% and high LDH levels in 42% of the patients. The following schedule was used: oral TMZ 100 mg/m2 on days 1 and 2 and FM iv 100 mg/m2 on day 2, 4 h after TMZ; A translational study aiming to analyse MGMT methylation status and base-excision repair (BER) gene expression was performed in a subset of 14 patients. RESULTS: We reported an overall response rate of 30.3% with 3 complete responses and a disease control rate of 50.5%. The related toxicity rate was low and mainly of haematological types. Although our population had a very poor prognosis, we observed a PFS of 6 months and an OS of 10 months. A non-significant correlation with response was found with the mean expression level of the three genes involved in the BER pathway (APE1, XRCC1 and PARP1), whereas no association was found with MGMT methylation status. CONCLUSION: This schedule could represent a good alternative for patients who are not eligible for immune or targeted therapy or whose previous therapies have failed. TRIAL REGISTRATION: EUDRACT 2009-016487-36l ; date of registration 23 June 2010.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Neoplasms/drug therapy , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Brain Neoplasms/secondary , DNA Modification Methylases/antagonists & inhibitors , DNA Repair Enzymes/antagonists & inhibitors , Drug Administration Schedule , Drug Resistance, Neoplasm/drug effects , Female , Humans , Male , Melanoma/mortality , Melanoma/secondary , Middle Aged , Nitrosourea Compounds/administration & dosage , Organophosphorus Compounds/administration & dosage , Prognosis , Progression-Free Survival , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Temozolomide/administration & dosage , Treatment Outcome , Tumor Suppressor Proteins/antagonists & inhibitors , Young Adult
4.
J Neurooncol ; 140(2): 427-434, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30109672

ABSTRACT

OBJECTIVE: This prospective, randomized, controlled and open-label clinical trial sought to evaluate the tolerability and efficacy of the FTD regimen (fotemustine, teniposide and dexamethasone) compared to HD-MA therapy (high-dose methotrexate plus cytarabine) and to elucidate some biomarkers that influence outcomes in patients with newly diagnosed primary CNS lymphoma. METHODS: Participants were stratified by IELSG risk score (low versus intermediate versus high) and randomly assigned (1:1) to receive four cycles of FTD or HD-MA regimen. Both regimens were administered every 3 weeks and were followed by whole-brain radiotherapy. The primary endpoints were overall response rate (ORR), progression-free survival (PFS) and overall survival (OS). RESULTS: Between June 2012, and June 2015, 52 patients were enrolled, of whom 49 patients were randomly assigned and analyzed. Of the 49 eligible patients, no significant difference was observed in terms of ORR between FTD (n = 24) and HD-MA (n = 25) groups (88% versus 84%, respectively, P = 0.628). Neither the 2-year PFS nor the 3-year OS rate differed significantly between FTD and HD-MA groups (37% versus 39% for 2-year PFS, P = 0.984; 51% versus 46% for 3-year OS, P = 0.509; respectively). The HD-MA group showed more serious neutropenia (P = 0.009) than the FTD group. High Bcl-6 expression correlated with longer OS (P = 0.038). CONCLUSIONS: FTD chemotherapy appeared to be safe and effective for PCNSL patients. High Bcl-6 expression correlated with longer survival.


Subject(s)
Antineoplastic Agents/administration & dosage , Central Nervous System Neoplasms/drug therapy , Lymphoma/drug therapy , Adolescent , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols , Biomarkers, Tumor/metabolism , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/mortality , Cytarabine/administration & dosage , Cytarabine/adverse effects , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Female , Humans , Lymphoma/metabolism , Lymphoma/mortality , Male , Methotrexate/administration & dosage , Methotrexate/adverse effects , Middle Aged , Nitrosourea Compounds/administration & dosage , Nitrosourea Compounds/adverse effects , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/adverse effects , Proto-Oncogene Proteins c-bcl-6/metabolism , Teniposide/administration & dosage , Teniposide/adverse effects , Treatment Outcome , Young Adult
5.
Br J Haematol ; 172(1): 111-21, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26458240

ABSTRACT

High-dose chemotherapy (HDT) with autologous stem cell transplantation is the standard of care for relapsed/refractory (RR) Hodgkin lymphoma (HL). Given that HDT may cure a sizeable proportion of patients refractory to first salvage, development of newer conditioning regimens remains a priority. We present the results of a novel HDT regimen in which carmustine was substituted by a third-generation chloroethylnitrosourea, fotemustine, with improved pharmacokinetics and safety (FEAM; fotemustine, etoposide, cytarabine, melphalan) in 122 patients with RR-HL accrued into a prospective registry-based study. Application of FEAM resulted in a 2-year progression-free survival (PFS) of 73·8% [95% confidence interval (CI), 0·64-0·81] with median PFS, overall survival and time to progression yet to be reached. The 2-year risk of progression adjusted for the competitive risk of death was 19·4% (95% CI, 0·12-0·27) for the entire patient population. Most previously established independent risk factors, except for fluorodeoxyglucose ((18) (F) FDG)-uptake, were unable to predict for disease progression and survival after FEAM. Although 32% of patients had (18) (F) FDG-positrin emission tomography-positive lesions before HDT, the 2-year risk of progression adjusted for competitive risk of death was 19·4% (95% CI; 0·12-0·27). No unusual acute toxicities or early/late pulmonary adverse events were registered. FEAM emerges as an ideal HDT regimen for RR-HL patients typically pre-exposed to lung-damaging treatments.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hodgkin Disease/drug therapy , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Cytarabine/administration & dosage , Cytarabine/adverse effects , Drug Evaluation/methods , Etoposide/administration & dosage , Etoposide/adverse effects , Female , Graft Survival , Hematopoietic Stem Cell Transplantation/methods , Hodgkin Disease/diagnostic imaging , Hodgkin Disease/therapy , Humans , Kaplan-Meier Estimate , Male , Melphalan/administration & dosage , Melphalan/adverse effects , Middle Aged , Nitrosourea Compounds/administration & dosage , Nitrosourea Compounds/adverse effects , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/adverse effects , Positron-Emission Tomography , Prospective Studies , Registries , Salvage Therapy/adverse effects , Salvage Therapy/methods , Transplantation Conditioning/methods , Treatment Outcome , Young Adult
6.
J Gene Med ; 18(9): 220-33, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27461566

ABSTRACT

BACKGROUND: The combination of stem cell-based gene therapy with chemotherapy comprises an advantageous strategy that results in a reduction of system toxicity effects and an improvement in the general efficacy of treatment. In the present study, we estimated the efficacy of adipose tissue-derived mesenchymal stem cells (AT-MSCs) expressing cytosine deaminase (CDA) combined with lysomustine chemotherapy in mice bearing late stage Lewis lung carcinoma (LLC). METHODS: Adipose tissue-derived mesenchymal stem cells were transfected with non-insert plasmid construct transiently expressing fused cytosine deaminase-uracil phosphoribosyltransferase protein (CDA/UPRT) or the same construct fused with Herpes Simplex Virus Type1 tegument protein VP22 (CDA/UPRT/VP22). Systemic administration of 5-fluorocytosine (5FC) and lysomustine was implemented after a single intratumoral injection of transfected AT-MSCs. RESULTS: We demonstrated that direct intratumoral transplantation of AT-MSCs expressing CDA/UPRT or CDA/UPRT/VP22 followed by systemic administration of 5FC resulted in a significant tumor growth inhibition. There was a 56% reduction in tumor volume in mice treated by AT-MSCs-CDA/UPRT + 5FC or with AT-MSCs-CDA/UPRT/VP22 + 5FC compared to control animals grafted with lung carcinoma alone. Transplantation of AT-MSCs-CDA/UPRT + 5FC and AT-MSCs-CDA/UPRT/VP22 + 5FC prolonged the life span of mice bearing LLC by 27% and 31%, respectively. Co-administration of lysomustine and AT-MSCs-CDA/UPRT + 5FC led to tumor growth inhibition (by 86%) and life span extension (by 60%) compared to the control group. CONCLUSIONS: Our data indicate that a combination CDA/UPRT-expressing AT-MSCs with lysomustine has a superior antitumor effect in the murine lung carcinoma model compared to monotherapies with transfected AT-MSCs or lysomustine alone, possibly because of a synergistic effect of the combination therapy. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Lewis Lung/therapy , Cytosine Deaminase/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Nitrosourea Compounds/pharmacology , Adipose Tissue/cytology , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Combined Modality Therapy , Cytosine Deaminase/genetics , Female , Flucytosine/administration & dosage , Flucytosine/pharmacology , Genetic Therapy/methods , Male , Mice, Inbred C57BL , Nitrosourea Compounds/administration & dosage , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Survival Analysis , Tumor Burden/drug effects , Tumor Burden/genetics
7.
Ann Hematol ; 95(9): 1513-9, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27365141

ABSTRACT

The BEAM regimen consisting of carmustine (BCNU), etoposide, cytarabine, and melphalan (MEL) is widely used before autologous hematopoietic stem cell transplantation (auto-HSCT) for lymphoma. However, intravenous BCNU is not available in Japan, and therefore, ranimustine (MCNU) has been used instead of BCNU (the MEAM regimen). We retrospectively analyzed the outcome of 79 adult patients who underwent auto-HSCT for lymphoma using this regimen in two centers, with 1- and 2-day dosing of MEL, respectively. Three-year overall survival (OS) and progression-free survival (PFS) probabilities were 77.3 and 56.5 % in the entire population and 71.7 and 58.0 % in patients with diffuse large B cell lymphoma. These outcomes were at least equivalent to those with the BEAM regimen. There was no regimen-related pulmonary toxicity. In a multivariate analysis, older age was the only factor that was significantly associated with for OS. In a comparison of the two MEL dosing schedules, while there was no significant differences in either OS or PFS, diarrhea was observed more frequently with 1-day dosing of MEL. In conclusion, the MEAM regimen appeared to be a promising conditioning regimen in auto-HSCT for lymphoma. A large prospective study is warranted to confirm the current findings.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma/therapy , Peripheral Blood Stem Cell Transplantation/methods , Transplantation Conditioning/methods , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cytarabine/administration & dosage , Cytarabine/adverse effects , Disease-Free Survival , Dose-Response Relationship, Drug , Etoposide/administration & dosage , Etoposide/adverse effects , Febrile Neutropenia/chemically induced , Female , Humans , Lymphoma/classification , Male , Melphalan/administration & dosage , Melphalan/adverse effects , Middle Aged , Nausea/chemically induced , Nitrosourea Compounds/administration & dosage , Nitrosourea Compounds/adverse effects , Retrospective Studies , Transplantation Conditioning/adverse effects , Transplantation, Autologous , Treatment Outcome , Young Adult
8.
J Neurooncol ; 128(3): 481-6, 2016 07.
Article in English | MEDLINE | ID: mdl-27165580

ABSTRACT

The optimal treatment of recurrent glioblastoma (GBM) in elderly patients is unclear. Fotemustine (FTM) is a third-generation nitrosourea showing efficacy in gliomas and it has been used with different schedules in adult patients. We performed, for the first time anywhere, a mono-institutional retrospective study to analyze the clinical outcome of an alternative fotemustine schedule in elderly patients with recurrent GBM. Retrospectively, we analyzed all GBM patients 65 years or older previously treated with the combination of radiation therapy and temozolomide (TMZ), receiving an alternative FTM schedule as second-line treatment at our Oncological Center from October 2011 to October 2014 with an ECOG PS ≤ 2. FTM was administrated at 80 mg/m(2) every 2 weeks for five consecutive administrations (induction phase), and then every 4 weeks at 80 mg/m(2) as maintenance. We enrolled 44 patients, 33 males and 11 females; average age was 70 years. ECOG PS was 0-1 in 80 % of the patients. 38 patients relapsed during temozolomide (TMZ) therapy. MGMT methylation status was analyzed in 34 patients and MGMT was methylated in 53 % of the patients. The median progression free survival (PFS) and overall survival (OS) from FTM treatment was 4.1 months (95 % CI 3.1-5.2) and 7 months (95 % CI 5.2-8.4), respectively. Patients with MGMT methylated status and patients who relapsed after completing TMZ therapy had a longer PFS and OS from the beginning of FTM. Thrombocytopenia was the most frequent grade 3-4 haematological toxicity (9 %). The alternative schedule of FTM may be an active and safe treatment for elderly patients with recurrent glioblastoma, especially patients with methylated MGMT and who relapsed after completing temozolomide therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Nitrosourea Compounds/administration & dosage , Organophosphorus Compounds/administration & dosage , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Drug Administration Schedule , Female , Humans , Male , Neoplasm Recurrence, Local/genetics , Nitrosourea Compounds/adverse effects , Organophosphorus Compounds/adverse effects , Recurrence , Retrospective Studies , Survival Analysis , Temozolomide , Treatment Outcome , Tumor Suppressor Proteins/genetics
9.
Ann Oncol ; 26(4): 798-803, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25538176

ABSTRACT

BACKGROUND: In the NIBIT-M1 study, we reported a promising activity of ipilimumab combined with fotemustine in metastatic melanoma (MM) patients with or without brain metastases. To corroborate these initial findings, we now investigated the long-term efficacy of this combination. PATIENTS AND METHODS: This analysis captured the 3-year outcome of MM patients who received ipilimumab combined with fotemustine as first- or second-line treatment. Median overall survival (OS), 3-year survival rates, immune-related (ir) progression-free survival (irPFS), brain PFS, and ir duration of response (irDOR) for the entire population and for patients with brain metastases were assessed. Clinical results were correlated with circulating CD3(+)CD4(+)ICOS(+)CD45RO(+) or CD45RA(+) T cells, neutrophil/lymphocyte (N/L) ratios, and tumorBRAF-V600 mutational status. RESULTS: Eighty-six MM patients, including 20 with asymptomatic brain metastases that had been pre-treated with radiotherapy in 7 subjects, were enrolled in the study. With a median follow-up of 39.9 months, median OS and 3-year survival rates were 12.9 months [95% confidence interval (CI) 7.1-18.7 months] and 28.5% for the whole study population, and 12.7 months (95% CI 2.7-22.7 months) and 27.8% for patients with brain metastases, respectively. Long-term ir adverse events consisting of G1 rush and pruritus occurred in 21% of patients. The absolute increase from baseline to week 12 in 'memory' but not in 'naïve' T cells identified patients with a better survival (P = 0.002). The N/L ratio correlated with a significantly better survival at early time points. BRAF status did not correlate with clinical outcome. CONCLUSIONS: Long-term analysis of the NIBIT-M1 trial continues to demonstrate efficacy of ipilimumab combined with fotemustine in MM patients. Fotemustine does not seem to impair the immunologic activity of ipilimumab. EUDRACT NUMBER: 2010-019356-50. CINICALTRIALSGOV: NCT01654692.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biological Therapy/mortality , Brain Neoplasms/drug therapy , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal/administration & dosage , Brain Neoplasms/mortality , Brain Neoplasms/secondary , Female , Follow-Up Studies , Humans , Ipilimumab , Male , Melanoma/mortality , Melanoma/pathology , Middle Aged , Neoplasm Staging , Nitrosourea Compounds/administration & dosage , Organophosphorus Compounds/administration & dosage , Prognosis , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Survival Rate
10.
Ann Oncol ; 25(3): 742-746, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24510314

ABSTRACT

BACKGROUND: In uveal melanoma (UM) with metastatic disease limited to the liver, the effect of an intrahepatic treatment on survival is unknown. We investigated prospectively the efficacy and toxicity of hepatic intra-arterial (HIA) versus systemic (IV) fotemustine in patients with liver metastases from UM. PATIENTS AND METHODS: Patients were randomly assigned to receive either IV or HIA fotemustine at 100 mg/m(2) on days 1, 8, 15 (and 22 in HIA arm only) as induction, and after a 5-week rest period every 3 weeks as maintenance. Primary end point was overall survival (OS). Response rate (RR), progression-free survival (PFS) and safety were secondary end points. RESULTS: Accrual was stopped after randomization of 171 patients based on the results of a futility OS analysis. A total of 155 patients died and 16 were still alive [median follow-up 1.6 years (range 0.25-6 years)]. HIA did not improve OS (median 14.6 months) when compared with the IV arm (median 13.8 months), hazard ratio (HR) 1.09; 95% confidence interval (CI) 0.79-1.50, log-rank P = 0.59. However, there was a significant benefit on PFS for HIA compared with IV with a median of 4.5 versus 3.5 months, respectively (HR 0.62; 95% CI 0.45-0.84, log-rank P = 0.002). The 1-year PFS rate was 24% in the HIA arm versus 8% in the IV arm. An improved RR was seen in the HIA (10.5%) compared with IV treatment (2.4%). In the IV arm, the most frequent grade ≥3 toxicity was thrombocytopenia (42.1%) and neutropenia (62.6%), compared with 21.2% and 28.7% in the HIA arm. The main grade ≥3 toxicity related to HIA was catheter complications (12%) and liver toxicity (4.5%) apart from two toxic deaths. CONCLUSION: HIA treatment with fotemustine did not translate into an improved OS compared with IV treatment, despite better RR and PFS. Intrahepatic treatment should still be considered as experimental. EUDRACT NUMBER AND CLINICALTRIALSGOV IDENTIFIER: 2004-002245-12 and NCT00110123.


Subject(s)
Antineoplastic Agents/administration & dosage , Liver Neoplasms/drug therapy , Melanoma/drug therapy , Nitrosourea Compounds/administration & dosage , Organophosphorus Compounds/administration & dosage , Uveal Neoplasms/drug therapy , Administration, Intravenous , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Disease-Free Survival , Female , Hepatic Artery , Humans , Infusions, Intra-Arterial , Liver/pathology , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Male , Melanoma/mortality , Melanoma/pathology , Middle Aged , Nitrosourea Compounds/adverse effects , Nitrosourea Compounds/therapeutic use , Organophosphorus Compounds/adverse effects , Organophosphorus Compounds/therapeutic use , Prospective Studies , Survival Analysis , Uveal Neoplasms/mortality , Uveal Neoplasms/pathology , Young Adult
11.
J Neurooncol ; 117(1): 85-92, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24458956

ABSTRACT

Studies on the monoclonal VEGF-A antibody bevacizumab gave raise to questions regarding the lack of an overall survival benefit, the optimal timing in the disease course and potential combination and salvage therapies. We retrospectively assessed survival, radiological progression type on bevacizumab and efficacy of salvage therapies in 42 patients with recurrent malignant gliomas who received bevacizumab and nitrosourea sequentially. 15 patients received bevacizumab followed by nitrosourea at progression and 27 patients vice versa. Time to treatment failure, defined as time from initiation of one to failure of the other treatment, was similar in both groups (9.6 vs. 9.2 months, log rank p = 0.19). Progression-free survival on nitrosoureas was comparable in both groups, while progression-free survival on bevacizumab was longer in the group receiving bevacizumab first (5.3 vs. 4.1 months, log rank p = 0.03). Survival times were similar for patients with grade III (n = 9) and grade IV (n = 33) tumors. Progression-free survival on bevacizumab for patients developing contrast-enhancing T1 progression was longer than for patients who displayed a non-enhancing T2 progression. However, post-progression survival times after bevacizumab failure were not different. Earlier treatment with bevacizumab was not associated with better outcome in this series. The fact that earlier as compared to later bevacizumab treatment does not result in a different time to treatment failure highlights the challenge for first-line or recurrence trials with bevacizumab to demonstrate an overall survival benefit if crossover of bevacizumab-naïve patients after progression occurs.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Glioma/drug therapy , Nitrosourea Compounds/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols , Bevacizumab , Brain/drug effects , Brain/pathology , Brain Neoplasms/pathology , Disease-Free Survival , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioma/pathology , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Retrospective Studies , Salvage Therapy , Treatment Outcome , Young Adult
12.
Bull Exp Biol Med ; 156(4): 480-2, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24771432

ABSTRACT

The efficiency of photodynamic therapy with photosensitizer Tiosens (Russia) was evaluated in mono- and combined therapy of rats with malignant gliomas (glioblastoma 101/8, oligodendroglioma 14-4-9, and C6 glioma). The efficiency of photodynamic monotherapy was not high: the animals died from brain edema developing in tumor tissue and in the adjacent normal cerebral tissue. Pathomorphological studies of tumor tissue detected necrosis and apoptosis, destruction of vessels with hemorrhages, and vascular thrombosis. Combined therapy for malignant gliomas including Tiosens photodynamic therapy and subsequent temodal or lysomustine chemotherapy, was the most effective. In glioblastoma 101/8, combined therapy with lysomustine or temodal led to prolongation of the lifespan by 127 %; 62.5 and 50 % rats were cured, respectively; in oligodendroglioma 14-4-9, animal lifespan was prolonged by 80 and 60 %, with 43 and 45 % rats cured, respectively. Glioma C6 was least sensitive to therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Oligodendroglioma/drug therapy , Photochemotherapy , Animals , Animals, Outbred Strains , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Screening Assays, Antitumor , Humans , Indoles/administration & dosage , Neoplasm Transplantation , Nitrosourea Compounds/administration & dosage , Organometallic Compounds/administration & dosage , Photosensitizing Agents/administration & dosage , Rats, Wistar , Temozolomide
13.
Rinsho Ketsueki ; 55(4): 440-4, 2014 04.
Article in Japanese | MEDLINE | ID: mdl-24850455

ABSTRACT

A 79-year-old man had been followed up since July 2003 based on a diagnosis of essential thrombocythemia (ET). The patient visited our hospital after developing a high fever and rash in August 2010, and Sweet's syndrome was diagnosed based on skin biopsy results. The bone marrow aspirate showed features like those of myelodysplastic/myeloproliferative neoplasm (MDS/MPN, unclassifiable). Administration of metenolone and azacitidine was initiated in March and May 2011, respectively, but the rash associated with Sweet's syndrome showed exacerbation. Ranimustine was therefore administered starting in July 2011 to control the blood cell count, but the rash associated with Sweet's syndrome persisted. Combination therapy with lenalidomide was initiated in September 2012, and resulted in control of the blood cell count and marked improvement of Sweet's syndrome.


Subject(s)
Sweet Syndrome/drug therapy , Sweet Syndrome/etiology , Thalidomide/analogs & derivatives , Thrombocythemia, Essential/complications , Thrombocythemia, Essential/drug therapy , Aged , Blood Cell Count , Bone Marrow/pathology , Drug Therapy, Combination , Humans , Lenalidomide , Male , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/pathology , Myelodysplastic-Myeloproliferative Diseases/etiology , Myelodysplastic-Myeloproliferative Diseases/pathology , Nitrosourea Compounds/administration & dosage , Skin/pathology , Sweet Syndrome/diagnosis , Sweet Syndrome/pathology , Thalidomide/administration & dosage , Thrombocythemia, Essential/blood , Treatment Outcome
14.
J Transl Med ; 11: 38, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23402397

ABSTRACT

BACKGROUND: The effect of the addition of fotemustine and/or interferon (IFN) to standard therapy with dacarbazine alone in patients with advanced malignant melanoma was investigated in a multicenter, randomized 2x2 factorial design trial. METHODS: A total of 260 patients were randomly assigned to one of four treatment groups: (A) fotemustine and dacarbazine repeated on 3-week cycle; (B) same treatment as (A) plus IFN-α2b three times per week; (C) dacarbazine alone repeated on 3-week cycle; (D) same treatment as (C) plus IFN-α2b three times per week. Two comparisons were planned to assess the efficacy of fotemustine (groups A+B vs. C+D) and IFN-α2b (groups A+C vs. B+D). RESULTS: Addition of fotemustine did not significantly improve overall survival (OS) (p=0.28) or progression-free survival (PFS) (p=0.55); Hazard ratio (HR) for OS was 0.93 (95% CI 0.71-1.21). Similarly, addition of IFN-α2b did not improve OS (p=0.68) or PFS (p=0.65); HR for OS was 0.92 (95% CI 0.70-1.20). Overall response rate was not improved by the addition of either fotemustine (p=0.87) or IFN-α2b (p=0.57). The combination of all three drugs resulted in the highest occurrence of adverse events. CONCLUSIONS: No significant improvement in outcomes were observed with the addition of either fotemustine or IFN-α2b to dacarbazine. TRIAL REGISTRATION: ClinicalTrials.gov: NCT01359956.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Dacarbazine/administration & dosage , Humans , Interferon alpha-2 , Interferon-alpha/administration & dosage , Nitrosourea Compounds/administration & dosage , Organophosphorus Compounds/administration & dosage , Recombinant Proteins/administration & dosage , Survival Analysis
15.
Am J Hematol ; 88(2): 102-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23224960

ABSTRACT

Since multiple myeloma (MM) is still not-curable, the management of relapse remains challenging. Given the known efficacy of alkylating agents in MM, we conducted a phase I/II study to test a new three drug combination in which Fotemustine (Muphoran), an alkylating agent of nitrosurea family, was added to bortezomib + dexamethasone backbone (B-MuD) for the treatment of MM relapsed patients. Fotemustine was administered at two dose levels (80-100 mg/m² i.v.) on day 1. The original 21-day schedule was early amended for extra-hematological toxicity and a 35-day schedule was adopted (Bortezomib 1.3 mg/m² i.v. on days 1, 8, 15, and 22, Dexamethasone 20 mg i.v. on days 1, 8, 15, and 22) for a total of six courses. Twenty-four patients were enrolled. The maximum tolerated dose of Fotemustine was 100 mg/m². The overall response rate was of 62% (CR 8%, VGPR 33%, and PR 21%). The median OS was 28.5 months, the median progression-free survival (PFS) was 19.1 months. B-MuD resulted effective in patients previous exposed to bortezomib without difference of response (P = 0.25) and PFS (P = 0.87) when compared to bortezomib-naive patients. Thrombocytopenia was the most common AE overall. In conclusion, B-MuD is an effective and well tolerated combination in relapsed MM patients even in advanced disease phase.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/therapeutic use , Dexamethasone/therapeutic use , Multiple Myeloma/prevention & control , Nitrosourea Compounds/therapeutic use , Organophosphorus Compounds/therapeutic use , Pyrazines/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Boronic Acids/administration & dosage , Boronic Acids/adverse effects , Bortezomib , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Neoplasm Staging , Nitrosourea Compounds/administration & dosage , Nitrosourea Compounds/adverse effects , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/physiopathology , Pyrazines/administration & dosage , Pyrazines/adverse effects , Secondary Prevention , Severity of Illness Index , Survival Analysis , Thrombocytopenia/chemically induced , Thrombocytopenia/physiopathology
16.
Lancet Oncol ; 13(9): 879-86, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22894884

ABSTRACT

BACKGROUND: Ipilimumab improves survival of patients with metastatic melanoma, many of whom develop brain metastases. Chemotherapy-induced release of tumour antigens might amplify ipilimumab's antitumour activity. We aimed to investigate the efficacy and safety of ipilimumab plus fotemustine in patients with metastatic melanoma with or without asymptomatic brain metastases. METHODS: In our open-label, single-arm phase 2 trial, we enrolled patients 18 years or older with measurable, locally advanced, unresectable stage III or stage IV melanoma between July 6, 2010, and April 14, 2011. Eligible patients had a life expectancy of 16 weeks or more and an Eastern Cooperative Oncology Group performance status of 1 or less, and could have received a maximum of one previous line of chemotherapy. Participants received induction treatment of 10 mg/kg intravenous ipilimumab every 3 weeks to a total of four doses, and 100 mg/m(2) intravenous fotemustine weekly for 3 weeks and then every 3 weeks from week 9 to week 24. Patients with a confirmed clinical response were eligible for maintenance treatment from week 24, with ipilimumab every 12 weeks and fotemustine every 3 weeks. The primary endpoint was the proportion of patients with immune-related disease control as established with immune-related response criteria. Analyses were done per protocol. This trial is registered with EudraCT, number 2010-019356-50, and with ClinicalTrials.gov, number NCT01654692. FINDINGS: 86 patients were eligible for treatment, of whom 20 had asymptomatic brain metastases at baseline. 40 patients in the study population achieved disease control (46·5%, 95% CI 35·7-57·6), as did ten with brain metastases (50·0%, 27·2-72·8). 47 patients (55%) had grade 3 or 4 treatment-related adverse events, of which the most common was myelotoxicity (thrombocytopenia in 21 [24%] patients and neutropenia in 16 [19%]). The most common grade 3 or 4 immune-related adverse events were hepatic: 21 patients (24%) had grade 3 or 4 increases in concentrations of alanine aminotransferase or aspartate aminotransferase. INTERPRETATION: The combination of ipilimumab plus fotemustine has clinical activity in patients with metastatic melanoma, including those with brain metastases. FUNDING: Bristol-Myers Squibb.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/secondary , Melanoma/drug therapy , Melanoma/secondary , Nitrosourea Compounds/administration & dosage , Organophosphorus Compounds/administration & dosage , Skin Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Neoplasms/diagnosis , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Disease-Free Survival , Female , Hematologic Diseases/chemically induced , Humans , Ipilimumab , Male , Melanoma/diagnosis , Melanoma/pathology , Middle Aged , Neoplasm Grading , Neoplasm Staging , Skin Neoplasms/diagnosis , Skin Neoplasms/pathology
17.
Ann Dermatol Venereol ; 139(4): 277-81, 2012 Apr.
Article in French | MEDLINE | ID: mdl-22482481

ABSTRACT

BACKGROUND: Fotemustine is an alkylating cytostatic drug belonging to the nitrosourea family and is used in particular in the treatment of disseminated malignant melanoma. Herein, we report a case of interstitial lung disease associated with fotemustine. PATIENTS AND METHODS: An 81-year-old man treated with fotemustine for metastatic melanoma presented acute interstitial lung disease 20 days after a fourth course of fotemustine monotherapy. The condition regressed spontaneously, with the patient returning to the clinical, radiological and blood gas status that had preceded fotemustine treatment. After other potential aetiologies had been ruled out, acute fotemustine-induced lung toxicity was considered and this treatment was definitively withdrawn. DISCUSSION: Other cytostatic agents belonging to the nitrosourea family can cause similar pictures, with a number of cases of interstitial lung disease thus being ascribed to fotemustine and dacarbazine. To our knowledge, this is the first case of interstitial lung disease induced by fotemustine monotherapy. This diagnosis should be considered where respiratory signs appear in melanoma patients undergoing fotemustine treatment.


Subject(s)
Antineoplastic Agents/adverse effects , Lung Diseases, Interstitial/chemically induced , Nitrosourea Compounds/adverse effects , Organophosphorus Compounds/adverse effects , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Humans , Male , Melanoma/drug therapy , Nitrosourea Compounds/administration & dosage , Organophosphorus Compounds/administration & dosage , Skin Neoplasms/drug therapy
19.
J Neurooncol ; 102(3): 417-24, 2011 May.
Article in English | MEDLINE | ID: mdl-20694830

ABSTRACT

In the present study we investigated the feasibility and effectiveness of a new biweekly schedule of fotemustine (FTM) in patients with recurrent glioblastoma, after at least one previous treatment. The primary endpoint was progression-free survival at 6 months; secondary objectives were clinical response, overall survival, disease-free survival, and toxicity. Forty patients (median age 52.8 years; median Karnofsky Performance Status at progression 90) underwent second-line chemotherapy with FTM. Selected patients were previously treated with a standard radiotherapy course with concomitant temozolomide (TMZ). After tumor relapse or progression proven by magnetic resonance imaging (MRI), all patients underwent chemotherapy with FTM, given intravenously at dose of 80 mg/m(2) every 2 weeks for five consecutive administrations (induction phase), and then every 4 weeks at 80 [DOSAGE ERROR CORRECTED] mg/m(2) as maintenance. A total of 329 infusions were administered; the median number of cycles administered was 8. All patients completed the induction phase, and 29 patients received at least one maintenance infusion. Response to treatment was assessed using MacDonald criteria. One complete response [2.5%, 95% confidence interval (CI): 0-10%], 9 partial responses (22.5%, 95% CI: 15-37%), and 16 stable diseases (40%, 95% CI: 32-51%) were observed. Median time to progression was 6.7 months (95% CI: 3.9-9.1 months). Progression-free survival at 6 months was 61%. Median survival from beginning of FTM chemotherapy was 11.1 months. The schedule was generally well tolerated; the main toxicities were hematologic (grade 3 thrombocytopenia in two cases). To the best of our knowledge, this is the first report specifically dealing with the use of a biweekly induction schedule of FTM. The study demonstrates that FTM has therapeutic efficacy as single-drug second-line chemotherapy with a favorable safety profile.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/prevention & control , Nitrosourea Compounds/administration & dosage , Organophosphorus Compounds/administration & dosage , Adult , Aged , Brain Neoplasms/mortality , Confidence Intervals , Dacarbazine/therapeutic use , Disease-Free Survival , Drug Administration Schedule , Female , Follow-Up Studies , Glioblastoma/mortality , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Odds Ratio , Temozolomide , Time Factors , Tomography, X-Ray Computed/methods
20.
Clin Cancer Res ; 27(17): 4737-4745, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34112708

ABSTRACT

PURPOSE: Phase II trials have shown encouraging activity with ipilimumab plus fotemustine and ipilimumab plus nivolumab in melanoma brain metastases. We report the primary analysis and 4-year follow-up of the NIBIT-M2 study, the first phase III trial comparing these regimens with fotemustine in patients with melanoma with brain metastases. PATIENTS AND METHODS: This phase III study recruited patients 18 years of age and older with BRAF wild-type or mutant melanoma, and active, untreated, asymptomatic brain metastases from nine centers, randomized (1:1:1) to fotemustine, ipilimumab plus fotemustine, or ipilimumab plus nivolumab. The primary endpoint was overall survival (OS). RESULTS: From January, 2013 to September, 2018, 27, 26, and 27 patients received fotemustine, ipilimumab plus fotemustine, and ipilimumab plus nivolumab. Median OS was 8.5 months [95% confidence interval (CI), 4.8-12.2] in the fotemustine arm, 8.2 months (95% CI, 2.2-14.3) in the ipilimumab plus fotemustine arm (HR vs. fotemustine, 1.09; 95% CI, 0.59-1.99; P = 0.78), and 29.2 months (95% CI, 0-65.1) in the ipilimumab plus nivolumab arm (HR vs. fotemustine, 0.44; 95% CI, 0.22-0.87; P = 0.017). Four-year survival rate was significantly higher for ipilimumab plus nivolumab than fotemustine [(41.0%; 95% CI, 20.6-61.4) vs. 10.9% (95% CI, 0-24.4; P = 0.015)], and was 10.3% (95% CI, 0-22.6) for ipilimumab plus fotemustine. In the fotemustine, ipilimumab plus fotemustine, and ipilimumab plus nivolumab arms, respectively, 11 (48%), 18 (69%), and eight (30%) patients had treatment-related grade 3 or 4 adverse events, without treatment-related deaths. CONCLUSIONS: Compared with fotemustine, ipilimumab plus nivolumab significantly improved overall and long-term survival of patients with melanoma with asymptomatic brain metastases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Ipilimumab/administration & dosage , Melanoma/drug therapy , Melanoma/secondary , Nitrosourea Compounds/administration & dosage , Nivolumab/administration & dosage , Organophosphorus Compounds/administration & dosage , Skin Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Female , Follow-Up Studies , Humans , Male , Melanoma/mortality , Middle Aged , Skin Neoplasms/mortality , Survival Rate , Time Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL