2,224 results on '"Antineoplastic Agents, Alkylating adverse effects"'
Search Results
2. Herpes zoster in patients with glioma treated with temozolomide.
- Author
-
Hiong A, Lapidus AH, Gately L, Sinclair G, and Ameratunga M
- Subjects
- Humans, Middle Aged, Male, Female, Retrospective Studies, Aged, Adult, Incidence, Risk Factors, Adrenal Cortex Hormones therapeutic use, Temozolomide therapeutic use, Herpes Zoster epidemiology, Glioma drug therapy, Glioma epidemiology, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Brain Neoplasms drug therapy, Brain Neoplasms epidemiology
- Abstract
Background: The risk of herpes zoster in patients treated with temozolomide is poorly defined in the literature. We aimed to evaluate the incidence of and risk factors for herpes zoster in individuals receiving temozolomide for glioma., Methods: A retrospective observational study was conducted on a series of patients treated with temozolomide for glioma at a single centre between 1 October 2018 and 30 September 2023., Results: 131 patients were treated with temozolomide for glioma with a median age of 55 years. 4 out of 131 patients (3.1 %) developed herpes zoster during temozolomide treatment. All cases of herpes zoster occurred in patients who had lymphocyte nadirs of less than 0.7 x 10
9 /L and were receiving corticosteroids concomitantly. The estimated herpes zoster incidence rates were 45.44 per 1000 person-years (95 % confidence interval (CI) 12.38-116.34 per 1000 person-years) in the overall study population and 224.97 per 1000 person-years (95 % CI 61.30-576.02 per 1000 person-years) in subjects who were treated with corticosteroids and had a lymphocyte nadir of less than 1.0 x 109 /L., Conclusion: Use of temozolomide, particularly in conjunction with lymphopaenia or corticosteroid use, poses a risk of herpes zoster. Further research into the benefits of prophylactic antiviral measures in this population is recommended., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Ltd. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
3. Evidence of apoptosis as an early event leading to cyclophosphamide-induced primordial follicle depletion in a prepubertal mouse model.
- Author
-
Hao X, Reyes Palomares A, Anastácio A, Liu K, and Rodriguez-Wallberg KA
- Subjects
- Animals, Female, Mice, Antineoplastic Agents, Alkylating pharmacology, Antineoplastic Agents, Alkylating toxicity, Antineoplastic Agents, Alkylating adverse effects, DNA Damage drug effects, Sexual Maturation drug effects, Cyclophosphamide pharmacology, Cyclophosphamide toxicity, Cyclophosphamide adverse effects, Apoptosis drug effects, Ovarian Follicle drug effects, Ovarian Follicle metabolism
- Abstract
Introduction: The mechanisms leading to ovarian primordial follicle depletion following gonadotoxic chemotherapy with cyclophosphamide and other cytotoxic drugs are currently understood through two main explanatory theories: apoptosis and over-activation. Discrepancies between the findings of different studies investigating these mechanisms do not allow to reach a firm conclusion. The heterogeneity of cell types in ovaries and their different degrees of sensitivity to damage, cell-cell interactions, periodical follicle profile differences, model age-dependent differences, and differences of exposure durations of tested drugs may partially explain the discrepancies among studies., Methods: This study used intact prepubertal mice ovaries in culture as study model, in which most follicles are primordial follicles. Histological and transcriptional analyses of ovaries exposed to the active metabolite of cyclophosphamide 4-hydroperoxycyclophosphamide (4-HC) were carried out via a time-course experiment at 8, 24, 48, and 72 h., Results: 4-HC treated ovaries showed a significant decrease in primordial follicle density at 24 h, along with active DNA damage (TUNEL) and overexpressed apoptosis signals (cleaved-poly ADP ribose polymerase in immunohistochemistry and western blotting). Meanwhile 4-HC treatment significantly up-regulated H2ax, Casp 6, Casp 8, Noxa, and Bax in ovaries, and up-regulated Puma in primordial follicles (FISH)., Discussion: Our results indicated that cyclophosphamide-induced acute ovarian primordial follicle depletion was mainly related to apoptotic pathways. No evidence of follicle activation was found, neither through changes in the expression of related genes to follicle activation nor in the density of growing follicles. Further validation at protein level in 4-HC-treated prepubertal mice ovaries at 24 h confirmed these observations., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Hao, Reyes Palomares, Anastácio, Liu and Rodriguez-Wallberg.)
- Published
- 2024
- Full Text
- View/download PDF
4. Risk and impact of cytomegalovirus infection in lymphoma patients treated with bendamustine.
- Author
-
Huang JP, Yeh CM, Gong YW, Tsai MH, Lin YT, Tsai CK, and Liu CJ
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Retrospective Studies, Risk Factors, Adult, Aged, 80 and over, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating therapeutic use, Incidence, Taiwan epidemiology, Cytomegalovirus, Bendamustine Hydrochloride therapeutic use, Bendamustine Hydrochloride adverse effects, Cytomegalovirus Infections epidemiology, Lymphoma drug therapy
- Abstract
Bendamustine is used to treat lymphoma with excellent efficacy but is known for its immunosuppressive effect. Cytomegalovirus (CMV) reactivation after bendamustine use has been reported. We aim to address the impact of CMV infection in lymphoma patients treated with bendamustine-containing regimens. We retrospectively analyzed lymphoma patients at Taipei Veterans General Hospital in Taiwan between September 1, 2010, and April 30, 2022. Clinically significant CMV infection (CS-CMVi) was defined as the first CMV reactivation after bendamustine use necessitating CMV therapy. Patients' baseline characteristics and laboratory data were recorded. The primary endpoint of the study was CS-CMVi. A time-dependent covariate Cox regression model was used to estimate the risk factors of CS-CMVi and mortality. A total of 211 lymphoma patients treated with bendamustine were enrolled. Twenty-seven (12.8%) had CS-CMVi. The cumulative incidence was 10.1 per 100 person-years during the three-year follow-up period. In the multivariate analysis, lines of therapy before bendamustine ≥ 1 (95% CI 1.10-24.76), serum albumin < 3.5 g/dL (95% CI 2.63-52.93), and liver disease (95% CI 1.51-28.61) were risk factors for CS-CMVi. In conclusion, CS-CMVi (95% confidence interval [CI] 1.23-10.73) was one of the major independent risk factors of mortality. Lines of therapy before bendamustine ≥ 1, hypoalbuminemia, and liver disease were risk factors for CS-CMVi in lymphoma patients treated with bendamustine., (© 2024. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
5. Real-life clinical benefit of oral metronomic cyclophosphamide administration in elderly and heavily pretreated epithelial ovarian cancer patients.
- Author
-
Attianese D, Massobrio R, Giorgi M, Villa M, Fuso L, Badellino E, Bellero M, and Ferrero A
- Subjects
- Humans, Female, Aged, Retrospective Studies, Middle Aged, Aged, 80 and over, Administration, Oral, CA-125 Antigen blood, Treatment Outcome, Cyclophosphamide administration & dosage, Cyclophosphamide adverse effects, Carcinoma, Ovarian Epithelial drug therapy, Ovarian Neoplasms drug therapy, Administration, Metronomic, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Agents, Alkylating adverse effects, Neoplasm Recurrence, Local drug therapy
- Abstract
Purpose: Oral metronomic cyclophosphamide (OMC) implicates the daily administration of low doses of chemotherapy. Its antitumor activity combined with an oral administration route and a good toxicity profile makes OMC an attractive option for heavily pretreated patients. We retrospectively evaluated OMC's clinical benefit and objective response in recurrent ovarian cancer patients., Methods: This is a retrospective observational study involving patients treated with OMC (50 mg daily) from 2017 to 2022 at the Academic Division Gynaecology, Mauriziano Hospital, Torino, Italy. Clinical benefit assessment included CA125 response, radiological response, and reported symptomatic improvement. Toxicities were reported using Common Terminology Criteria for Adverse Events version 5.0., Results: Thirty-eight patients (average age 72, range 49-88) were included. 90% had FIGO stage III/IV at diagnosis and 64% underwent ≥ 3 previous lines of chemotherapy. Before OMC, 79% had ECOG 1 or 2. 8.6% of patients had a partial response (PR), and 40% a stable disease (SD). Median duration of response was 7.4 months. After 3 months on OMC, 51% experienced symptom improvement, and 53.3% experienced Ca125 reduction or stabilization. 66.7% of patients older than 75 responded to treatment; in 40% of cases, responses lasted ≥ 6 months (p = 0.08). No G3-4 hematological toxicities occurred. Nausea and fatigue G1-G2 were reported in 5 (13%) and 13 (34%) cases, respectively., Conclusion: OMC is a feasible therapeutic option for recurrent ovarian cancer, providing satisfying clinical responses with a good toxicity profile, even in elderly and heavily pretreated patients with a suboptimal performance status., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Ifosfamide-induced acute kidney injury and confusional syndrome in a patient with diffuse large B-cell lymphoma.
- Author
-
Lizondo López T, Rivero A, and Carcelero San Martín E
- Subjects
- Humans, Male, Aged, Middle Aged, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse complications, Acute Kidney Injury chemically induced, Ifosfamide adverse effects, Confusion chemically induced, Confusion etiology, Antineoplastic Agents, Alkylating adverse effects
- Published
- 2024
- Full Text
- View/download PDF
7. One Case of Abnormal Decrease of Interferon Gamma Release Assay Result Caused by Melphalan.
- Author
-
Li G, Lu T, and Shan N
- Subjects
- Humans, Male, Antineoplastic Agents, Alkylating adverse effects, Middle Aged, Bone Marrow drug effects, Bone Marrow pathology, Interferon-gamma blood, False Negative Reactions, Melphalan therapeutic use, Melphalan adverse effects, Multiple Myeloma drug therapy, Multiple Myeloma blood, Interferon-gamma Release Tests methods
- Abstract
Background: Interferon gamma release assay (IGRA) is an important method to detect the specific antigen of tuberculosis, which is crucial to the diagnosis of tuberculosis or potential tuberculosis infection., Methods: We report a case of myelosuppression caused by the use of Melphalan in the treatment of multiple myeloma, resulting in an abnormal decrease in interferon gamma release assay results., Results: We collected blood samples from the patient for retesting and the result of the test did not differ significantly. Upon reviewing the case, it was found that the patient's use of Melphalan treatment resulted in bone marrow suppression and extreme reduction of peripheral blood lymphocytes. Therefore, it is speculated that the abnormal decrease of the interferon gamma release assay result is caused by bone marrow suppression, which is caused by the use of Melphalan., Conclusions: When patients with multiple myeloma are treated with Melphalan, it can lead to bone marrow suppression and result in false negative interference gamma release assay results. Laboratory staff should consider the existence of such interference and communicate with clinical doctors in a timely manner.
- Published
- 2024
- Full Text
- View/download PDF
8. Perception of pneumocystis jirovecii pneumonia (PJP) prophylaxis in glioma patients receiving concurrent temozolomide and radiation- a patient and physician survey.
- Author
-
Beltran-Bless AA, Alshamsan B, Jia J, Lo V, Climans S, Nicholas G, and Ng TL
- Subjects
- Humans, Middle Aged, Male, Female, Adult, Aged, Young Adult, Surveys and Questionnaires, Physicians, Chemoradiotherapy adverse effects, Antibiotic Prophylaxis, Temozolomide therapeutic use, Glioma radiotherapy, Glioma complications, Pneumonia, Pneumocystis prevention & control, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Pneumocystis carinii, Brain Neoplasms radiotherapy
- Abstract
Purpose: Pneumocystis jirovecii pneumonia (PJP) prophylaxis is required by provincial and national drug monographs during glioma treatment using temozolomide (TMZ) concurrently with radiation (TMZ-RT). However, real-world data suggest the potential benefits of PJP prophylaxis may not outweigh its potential harms in this population., Methods: We conducted a single-center patient survey and a national physician survey to explore the role of PJP prophylaxis amongst glioma patients undergoing TMZ-RT., Results: 23% (31/133) of physicians and 60% (44/73) of patients completed a survey. The median patient age was 42 (range 20-77); 85% (34/40) had completed adjuvant TMZ. Although only 2.4% (1/41) of patients received PJP prophylaxis, only one person (without PJP prophylaxis) was hospitalized for pneumonia. When presented with hypothetical PJP risks, 13.2% (5/38) of patients were concerned about PJP infection, while 26% (10/38) were concerned about potential side effects from prophylactic antibiotics. Most physicians (77%, 17/22) perceived the evidence for PJP prophylaxis as weak; 58% (11/19) did not routinely prescribe prophylaxis, and 73% (16/22) felt that PJP prophylaxis should be limited to patients with additional risk factors. Over 95% of physicians estimated that the incidence of PJP was < 1% in their last 5 years of practice regardless of PJP prophylaxis. For 73% (16/22) of physicians, to prescribe PJP prophylaxis, the risk of PJP infection needed to be 3-8%., Conclusion: The current recommendation to routinely prescribe PJP prophylaxis in patients receiving TMZ-RT in the absence of other risk factors warrants reconsideration., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
9. Interindividual variation in ovarian reserve after gonadotoxic treatment in female childhood cancer survivors - a genome-wide association study: results from PanCareLIFE.
- Author
-
van der Perk MEM, Broer L, Yasui Y, Laven JSE, Robison LL, Tissing WJE, Versluys B, Bresters D, Kaspers GJL, Lambalk CB, Overbeek A, Loonen JJ, Beerendonk CCM, Byrne J, Berger C, Clemens E, van Dulmen-den Broeder E, Dirksen U, van der Pal HJ, de Vries ACH, Winther JF, Ranft A, Fosså SD, Grabow D, Muraca M, Kaiser M, Kepák T, Kruseova J, Modan-Moses D, Spix C, Zolk O, Kaatsch P, Kremer LCM, Brooke RJ, Wang F, Baedke JL, Uitterlinden AG, Bos AME, van Leeuwen FE, Ness KK, Hudson MM, van der Kooi ALF, and van den Heuvel-Eibrink MM
- Subjects
- Humans, Female, Adult, Young Adult, Antineoplastic Agents, Alkylating adverse effects, Neoplasms genetics, Neoplasms drug therapy, Risk Factors, Child, Adolescent, Europe epidemiology, Genome-Wide Association Study, Cancer Survivors, Ovarian Reserve genetics, Ovarian Reserve drug effects, Ovarian Reserve radiation effects, Anti-Mullerian Hormone blood, Anti-Mullerian Hormone genetics, Polymorphism, Single Nucleotide
- Abstract
Objective: To discover new variants associated with low ovarian reserve after gonadotoxic treatment among adult female childhood cancer survivors using a genome-wide association study approach., Design: Genome-wide association study., Setting: Not applicable., Patients: A discovery cohort of adult female childhood cancer survivors from the pan-European PanCareLIFE cohort (n = 743; median age: 25.8 years), excluding those who received bilateral ovarian irradiation, bilateral oophorectomy, central nervous system or total body irradiation, or stem cell transplantation. Replication was attempted in the US-based St. Jude Lifetime Cohort (n = 391; median age: 31.3 years)., Exposure: Female childhood cancer survivors are at risk of therapy-related gonadal impairment. Alkylating agents are well-established risk factors, and the interindividual variability in gonadotoxicity may be explained by genetic polymorphisms. Data were collected in real-life conditions, and cyclophosphamide equivalent doses were used to quantify alkylation agent exposure., Main Outcome Measure: Anti-Müllerian hormone (AMH) levels served as a proxy for ovarian function, and the findings were combined in a meta-analysis., Results: Three genome-wide significant (<5.0 × 10
-8 ) and 16 genome-wide suggestive (<5.0 × 10-6 ) loci were associated with log-transformed AMH levels, adjusted for cyclophosphamide equivalent dose of alkylating agents, age at diagnosis, and age at study in the PanCareLIFE cohort. On the basis of the effect allele frequency (EAF) (>0.01 if not genome-wide significant), and biologic relevance, 15 single nucleotide polymorphisms were selected for replication. None of the single nucleotide polymorphisms were statistically significantly associated with AMH levels. A meta-analysis indicated that rs78861946 was associated with borderline genome-wide statistical significance (reference/effect allele: C/T; effect allele frequency: 0.04, beta (SE): -0.484 (0.091)., Conclusion: This study found no genetic variants associated with a lower ovarian reserve after gonadotoxic treatment because the findings of this genome-wide association study were not statistically significant replicated in the replication cohort. Suggestive evidence for the potential importance of 1 variant is briefly discussed, but the lack of statistical significance calls for larger cohort sizes. Because the population of childhood cancer survivors is increasing, large-scale and systematic research is needed to identify genetic variants that could aid predictive risk models of gonadotoxicity as well as fertility preservation options for childhood cancer survivors., Competing Interests: Declaration of Interests M.E.M.v.d.P. reports funding from European Union’s Seventh Framework Programme for research (technological development, and demonstration funding), Dutch Cancer Society (grant no. VU 2006-3622), Princess Máxima Center Foundation, and Stichting Kinderoncologisch Centrum Rotterdam (sKOCR), DCOG-LATER VEVO study was funded by the Dutch Cancer Society (grant no. VU 2006-3622) and by the Children Cancer-Free Foundation (Project no. 20) and the St Jude Lifetime Cohort study by NCI U01 CA195547. L.B. has nothing to disclose. Y.Y. reports funding from National Cancer Institute (NCI) U01CA195547 (MPI: Hudson/Ness), St. Jude Lifetime Cohort Study (SJLIFE), NCI P30CA021765 (PI: Roberts) Cancer Center Support Grant (CCSG), and American Lebanese Syrian Associated Charities for the submitted work; funding from NCI U24CA055727 (PI: Armstrong) Childhood Cancer Survivor Study (CCSS), National Institutes of Health (NIH) R03DE029238 (PI: Li/Sun) Innovative Statistical Analysis for Genome-Wide Data with General Interval Censored Outcomes of Oral Health in Childhood Cancer Survivors, NCI R01CA216354 (MPI: Yasui/Zhang) Late Effects Prediction Using Clinical Phenotypes and Whole Genome Sequencing, St Jude Lifetime Cohort study by NCI U01 CA195547, NCI R01CA270157 (MPI: Bhakta/Yasui) Measuring Lifelong Multimorbidity with Patient Perspectives: Implementing and Visualizing the Cumulative Burden Methodology Across Cohorts, NCI R01CA258193 (MPI: Huang/Yasui) Patient-Generated Health Data to Predict Childhood Cancer Survivorship Outcomes (HEALTHSHARE), NIH &MD Anderson Cancer Center R01CA261750-01A1 (MPI: Howell/Mulrooney/Yasui) Personalized Risk Prediction to Reduce Cardiovascular Disease in Childhood Cancer Survivors, NCI &Northwestern University R01CA261898 (MPI: Burridge/Sapkota) Predicting and Preventing Chemotherapy-Induced Cardiotoxicity in African American Children, NC. J.S.E.L. has nothing to disclose. L.L.R. has nothing to disclose. W.J.E.T. has nothing to disclose. B.V. has nothing to disclose. B.V. has nothing to disclose. D.B. has nothing to disclose. G.J.L.K. has nothing to disclose. C.B.L. has nothing to disclose. A.O. has nothing to disclose. J.J.L. has nothing to disclose. C.C.M.B. has nothing to disclose. J.B. has nothing to disclose. C.B. has nothing to disclose. E.C. has nothing to disclose. E.v.D.-d.B. has nothing to disclose. U.D. has nothing to disclose. H.J.v.d.P. has nothing to disclose. A.C.H.d.V. has nothing to disclose. J.F.W. has nothing to disclose. A.R. has nothing to disclose. S.D.F. has nothing to disclose. D.G. has nothing to disclose. M.M. has nothing to disclose. M.K. has nothing to disclose. T.K. has nothing to disclose. J.K. has nothing to disclose. D.M.-M. has nothing to disclose. C.S. has nothing to disclose. O.Z. has nothing to disclose. P.K. has nothing to disclose. L.C.M.K. has nothing to disclose. R.J.B. has nothing to disclose. F.W. has nothing to disclose. J.L.B. has nothing to disclose. A.G.U. has nothing to disclose. A.M.E.B. has nothing to disclose. F.E.v.L. has nothing to disclose. K.K.N. has nothing to disclose. M.M.H. has nothing to disclose. A.-L.L.F.v.d.K. has nothing to disclose. M.M.v.d.H.E. has nothing to disclose., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
10. Molecular Iodine Improves the Efficacy and Reduces the Side Effects of Metronomic Cyclophosphamide Treatment against Mammary Cancer Progression.
- Author
-
Delgado-González E, Ríos-Arellano EL, Anguiano B, and Aceves C
- Subjects
- Animals, Female, Rats, Mammary Neoplasms, Experimental drug therapy, Mammary Neoplasms, Experimental pathology, Mammary Neoplasms, Experimental metabolism, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor A genetics, Disease Progression, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Agents, Alkylating pharmacology, Cyclophosphamide pharmacology, Cyclophosphamide adverse effects, Cyclophosphamide administration & dosage, Administration, Metronomic, Rats, Sprague-Dawley, Iodine administration & dosage, Iodine pharmacology
- Abstract
Metronomic chemotherapy with cyclophosphamide (Cpp) has shown promising results in cancer protocols. These lower and prolonged doses have antiangiogenic, pro-cytotoxic, and moderate secondary effects. Molecular iodine (I
2 ) reduces the viability of cancer cells and, with chemotherapeutic agents, activates the antitumoral immune response and diminishes side effects. The present work evaluates the adjuvant of oral I2 with Cpp using a murine model of mammary cancer. Female Sprague Dawley rats with 7,12-dimethylbenzantracene-induced tumors received Cpp intraperitoneal (50 and 70 mg/kg two times/week, iCpp50 and iCpp70) and oral (0.03%; 50 mg/Kg; oCpp50) doses. I2 (0.05%, 50 mg/100 mL) and oCpp50 were offered in drinking water for three weeks. iCpp70 was the most efficient antitumoral dose but generated severe body weight loss and hemorrhagic cystitis (HC). I2 prevented body weight loss, exhibited adjuvant actions with Cpp, decreasing tumor growth, and canceled HC mechanisms, including decreases in vascular endothelial growth factor (VEGF) and Survivin expression. oCpp50 + I2 diminished angiogenic signals (CD34, vessel-length, and VEGF content) and proinflammatory cytokines (interleukin-10 and tumor necrosis factor-alpha) and increased cytotoxic (lymphocytic infiltration, CD8+ cells, Tbet, and interferon-gamma) and antioxidant markers (nuclear erythroid factor-2 and glutathione peroxidase). I2 enhances the effectiveness of oCpp, making it a compelling candidate for a clinical protocol.- Published
- 2024
- Full Text
- View/download PDF
11. The protection afforded by kefir against cyclophosphamide induced testicular toxicity in rats by oxidant antioxidant and histopathological evaluations.
- Author
-
Cetik Yildiz S, Demir C, Cengiz M, Irmak H, Peker Cengiz B, and Ayhanci A
- Subjects
- Animals, Male, Rats, Oxidants metabolism, Antineoplastic Agents, Alkylating toxicity, Antineoplastic Agents, Alkylating adverse effects, Rats, Wistar, Cyclophosphamide toxicity, Cyclophosphamide adverse effects, Testis drug effects, Testis metabolism, Testis pathology, Antioxidants pharmacology, Antioxidants metabolism, Oxidative Stress drug effects, Kefir
- Abstract
Cyclophosphamide (CTX) is the most commonly used effective alkylating drug in cancer treatment, but its use is restricted because its toxic side effect causes testicular toxicity. CTX disrupts the tissue redox and antioxidant balance and the resulting tissue damage causes oxidative stress. In our study based on this problem, kefir against CTX-induced oxidative stress and testicular toxicity were investigated. Rats were divided into 6 groups: control, 150 mg/kg CTX, 5 and 10 mg/kg kefir, 5 and 10 mg/kg kefir + 150 CTX. While the fermented kefirs were mixed and given to the rats for 12 days, CTX was given as a single dose on the 12th day of the experiment. Testis was scored according to spermatid density, giant cell formation, cells shed into tubules, maturation disorder, and atrophy. According to our biochemical findings, the high levels of total oxidant status (TOS), and the low levels of total antioxidant status (TAS) in the CTX group, which are oxidative stress markers, indicate the toxic effect of CTX, while the decrease in TOS levels and the increase in TAS levels in the kefir groups indicate the protective effect of kefir. In the CTX-administered group, tubules with impaired maturation and no spermatids were observed in the transverse section of the testicle, while in the kefir groups, the presence of near-normal tubule structures and tubule lumens despite CTX showed the protective effect of kefir. In our study, it was observed that kefir had a protective and curative effect on CTX-induced toxicity and oxidative stress and could be a strong protector., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Effectiveness of trabectedin for radiation-induced angiosarcoma of the breast refractory to several anticancer drugs.
- Author
-
Matsuzaki E, Kajihara I, Tomiguchi M, Kimura T, Araki S, Sawamura S, Makino K, Aoi J, Masuguchi S, and Fukushima S
- Subjects
- Humans, Female, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating therapeutic use, Treatment Outcome, Middle Aged, Aged, Drug Resistance, Neoplasm, Hemangiosarcoma drug therapy, Hemangiosarcoma etiology, Hemangiosarcoma diagnosis, Breast Neoplasms radiotherapy, Breast Neoplasms drug therapy, Trabectedin administration & dosage, Trabectedin therapeutic use, Trabectedin adverse effects, Neoplasms, Radiation-Induced diagnosis, Neoplasms, Radiation-Induced etiology, Neoplasms, Radiation-Induced drug therapy
- Published
- 2024
- Full Text
- View/download PDF
13. Lomustine overdose in a patient with diffuse glioma: symptoms, management and outcome.
- Author
-
Heggebø LC, Blakstad H, Eriksson MC, and Brandal P
- Subjects
- Humans, Male, Pancytopenia chemically induced, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Agents, Alkylating adverse effects, Glioma drug therapy, Quality of Life, Middle Aged, Lomustine administration & dosage, Brain Neoplasms drug therapy, Drug Overdose, Oligodendroglioma drug therapy
- Abstract
A male patient started PCV chemotherapy (a combination of procarbazine, lomustine and vincristine) for a recurrent oligodendroglioma grade 2. Unfortunately, our patient took an unintended overdose of lomustine during the first PCV course: instead of 160 mg absolute dose of lomustine on day 1 only, he consumed 160 mg absolute dose of lomustine for seven consecutive days to a total dose of 1120 mg. Pancytopenia became evident after 24 days, and several months of severe myelosuppression, infections, reduced general condition, and nutrition difficulties followed. Fortunately, our patient with time recovered his bone marrow function. However, the patient's quality of life was reduced for a long time and several lessons were learnt: oral and written information on chemotherapy is essential, but not always sufficient to ensure the correct dosing of patient-administered chemotherapy. Oral chemotherapeutics should be delivered as a single-dose supply or be administered by experienced health personnel., Competing Interests: Competing interests: None declared., (© BMJ Publishing Group Limited 2024. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
14. Letter: Efficacy and Safety of Carmustine Wafer Implantation After Ventricular Opening in Glioblastomas, Isocitrate Dehydrogenase-Wildtype, in Adults.
- Author
-
Andrade-Andrade P, Ordóñez-Rubiano EG, and Hakim F
- Subjects
- Humans, Adult, Treatment Outcome, Glioblastoma drug therapy, Glioblastoma surgery, Brain Neoplasms surgery, Isocitrate Dehydrogenase genetics, Carmustine administration & dosage, Carmustine adverse effects, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating administration & dosage
- Published
- 2024
- Full Text
- View/download PDF
15. Clinical Course after Carmustine Wafer Implantation for Newly Diagnosed Adult-type Diffuse Gliomas; A controlled propensity matched analysis of a single center cohort.
- Author
-
Ono T, Suzuki H, Nanjo H, and Shimizu H
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Retrospective Studies, Prognosis, Cohort Studies, Aged, Drug Implants, Survival Rate, Follow-Up Studies, Carmustine administration & dosage, Carmustine adverse effects, Carmustine therapeutic use, Glioma drug therapy, Brain Neoplasms surgery, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating administration & dosage, Propensity Score
- Abstract
Purpose: It remains unclear whether combining carmustine wafer (CW) implantation with the standard treatment for adult-type diffuse gliomas is safe and has a prognostic impact. This study aimed to investigate the prognostic value and safety of CW implantation., Methods: Adult patients with IDH-wild-type and -mutant gliomas, grades 3-4 treated with surgical resection, radiotherapy, and temozolomide chemotherapy between 2013 and 2023 were surveyed. CWs were implanted except in cases of intraoperative wide ventricle opening or marked preoperative brain swelling. For survival analyses, a case-matched dataset based on propensity score matching (PSM), including multiple factors (patient background, diagnosis, and extent of resection) was generated. Progression-free survival (PFS), overall survival (OS), and frequency of complications of CW implantation (brain edema, infection, and cerebrospinal fluid leakage) were compared between the CW and non-use groups., Results: In total, 127 patients (75 in the CW use group and 52 in the non-use group) were enrolled. Regardless of stratification, no significant differences in PFS and OS were observed between the CW use and non-use groups. The frequency of postoperative brain edema was significantly higher in the CW use group than in the non-use group. An adjusted dataset containing 41 patients in the CW use and nonuse groups was generated. Even after PSM, CW implantation had no prognostic effect., Conclusions: CW implantation with standard treatment demonstrated little beneficial effect for the present strategy of CW use., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
16. Pleuroparenchymal fibroelastosis as a late complication of childhood cancer therapy: A case series.
- Author
-
Marathe PH, Santibanez V, Meyers PA, Padilla ML, and Friedman DN
- Subjects
- Adolescent, Child, Child, Preschool, Female, Humans, Male, Antineoplastic Agents, Alkylating adverse effects, Hematopoietic Stem Cell Transplantation adverse effects, Neoplasms drug therapy, Neoplasms complications, Neoplasms pathology, Pulmonary Fibrosis etiology, Pulmonary Fibrosis pathology, Lung Diseases, Interstitial pathology, Lung Diseases, Interstitial etiology, Lung Diseases, Interstitial chemically induced, Lung Diseases, Interstitial therapy
- Abstract
Pleuroparenchymal fibroelastosis (PPFE) is a rare interstitial pneumonia with distinct clinicopathologic features. It has been associated with exposure to hematopoietic stem cell transplantation (HSCT) and classical alkylating agents. Here, we highlight PPFE as a late complication of childhood cancer therapy by describing the cases of four survivors of childhood cancer with a diagnosis of treatment-related PPFE. All patients received high-dose alkylating agents. PPFE should be considered in the differential diagnosis of restrictive lung disease in patients with history of exposure to alkylating agents or HSCT. Development of PPFE-specific, noninvasive diagnostic tools and disease-modifying therapies will clinically benefit these patients., (© 2024 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
17. Correlation of Genetic Polymorphism of CYP3A5 to Cyclophosphamide Efficacy and Toxicity in Rhabdomyosarcoma Pediatric Egyptian Cancer Patients.
- Author
-
ElShereef CE, Zaki HF, Badary OA, Kamal S, Nagy M, Makhlouf D, Kamal M, Elnadi I, A Abdelshafi S, Abou El Naga S, and Saber MM
- Subjects
- Humans, Male, Female, Child, Egypt epidemiology, Child, Preschool, Retrospective Studies, Follow-Up Studies, Prognosis, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating therapeutic use, Infant, Genotype, Adolescent, Survival Rate, Cytochrome P-450 CYP3A genetics, Cyclophosphamide adverse effects, Rhabdomyosarcoma genetics, Rhabdomyosarcoma drug therapy, Polymorphism, Single Nucleotide
- Abstract
Objectives: Rhabdomyosarcoma (RMS) accounts for 50% of soft tissue sarcomas and 7% of pediatric malignancies. Cyclophosphamide (CPA) is the cornerstone of therapy and is a prodrug that is activated by the highly polymorphic drug-metabolizing enzyme CYP3A5. We aim to examine the possible CYP3A5 polymorphism association with CPA efficacy, survival outcomes, and toxicity in Egyptian pediatric RMS patients., Methods: The three non-functional SNPs, CYP3A5*3 rs776746 (C_26201809_30), CYP3A5*6 rs10264272 (C_30203950_10), and CYP3A5*7 rs41303343 (C_32287188_10) were genotyped by real-time PCR. We conducted a cohort retrospective study of 150 pediatric RMS patients treated with CPA-based first-line treatment to analyze the association between these genotypes and CPA efficacy/toxicities in RMS patients., Key Findings: The frequency of having normal, intermediate, and poor metabolizers was 4.7%, 34%, and 61.3%, respectively. There was an association between these different phenotypes, genotypes, and CPA efficacy/toxicity. Hemorrhagic cystitis and pancytopenia were present in all patients, while nephrotoxicity incidence was 87.3%. There was a notable difference in the occurrence of hemorrhagic cystitis among CYP3A5 intermediate metabolizers *1/*3, *1/*6, and poor metabolizers *3/*3, *3/*6 with a significance level of p<0.05. Neither CYP3A5*7 polymorphism nor *6/*6 genotype was identified in our study., Conclusion: Our results demonstrate that CYP3A5*3 (rs776746) and CYP3A5*6 (rs10264272) have a great association with CPA efficacy and toxicity in RMS patients.
- Published
- 2024
- Full Text
- View/download PDF
18. Infection risk and antimicrobial prophylaxis in bendamustine-treated patients with indolent non-Hodgkin lymphoma: An Australasian Lymphoma Alliance study.
- Author
-
Manos K, Churilov L, Grigg A, Di Ciaccio P, Wong J, Chandra Sekaran U, Wight J, Goh Z, Jina H, Butler L, Yannakou CK, Hamad N, Gregory GP, Gangatharan S, Cochrane T, Hawkes EA, and Lasica M
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Retrospective Studies, Adult, Aged, 80 and over, Antibiotic Prophylaxis methods, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating therapeutic use, Bendamustine Hydrochloride therapeutic use, Bendamustine Hydrochloride adverse effects, Bendamustine Hydrochloride administration & dosage, Lymphoma, Non-Hodgkin drug therapy, Lymphopenia chemically induced, Opportunistic Infections prevention & control
- Abstract
Infection and lymphopenia are established bendamustine-related complications. The relationship between lymphopenia severity and infection risk, and the role of antimicrobial prophylaxis, is not well described. This multicentre retrospective study analysed infection characteristics and antimicrobial prophylaxis in 302 bendamustine-treated indolent non-Hodgkin lymphoma patients. Lymphopenia (<1 × 10
9 /L) was near universal and time to lymphocyte recovery correlated with cumulative bendamustine dose. No association between lymphopenia severity and duration with infection was observed. Infections occurred in 44% of patients (50% bacterial) with 27% hospitalised; 32% of infections occurred ≥3 months post bendamustine completion. Infection was associated with obinutuzumab and/or maintenance anti-CD20 therapy, prior therapy and advanced stage. Twenty-four opportunistic infections occurred in 21 patients: ten varicella zoster virus (VZV), seven herpes simplex virus (HSV), one cytomegalovirus, one progressive multifocal leucoencephalopathy, one nocardiosis, one Pneumocystis jiroveci pneumonia (PJP) and three other fungal infections. VZV/HSV and PJP prophylaxis were prescribed to 42% and 54% respectively. Fewer VZV/HSV infections occurred in patients receiving prophylaxis (HR 0.14, p = 0.061) while PJP prophylaxis was associated with reduced risk of bacterial infection (HR 0.48, p = 0.004). Our study demonstrates a significant infection risk regardless of lymphopenia severity and supports prophylaxis to mitigate the risk of early and delayed infections., (© 2024 British Society for Haematology and John Wiley & Sons Ltd.)- Published
- 2024
- Full Text
- View/download PDF
19. Elderly long-term survivors in the Nordic phase II study with first-line maintenance temozolomide for primary central nervous system lymphoma: a 10-year follow-up.
- Author
-
Pulczynski EJ, Simonsen MR, Kuittinen O, Fagerli UM, Erlanson M, Fluge Ø, Leppä S, Østenstad B, Fosså A, Eriksson M, El-Galaly T, Kuitunen H, Papworth K, Ljungqvist M, Pedersen MB, and Pollari M
- Subjects
- Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Cancer Survivors, Dacarbazine therapeutic use, Dacarbazine administration & dosage, Follow-Up Studies, Lymphoma drug therapy, Lymphoma mortality, Maintenance Chemotherapy, Treatment Outcome, Central Nervous System Neoplasms drug therapy, Central Nervous System Neoplasms mortality, Temozolomide therapeutic use
- Published
- 2024
- Full Text
- View/download PDF
20. A retrospective, real-life analysis of metronomic oral single-agent cyclophosphamide for the treatment of platinum-pretreated advanced ovarian carcinoma in Italy.
- Author
-
Gebbia V, Martorana F, Scandurra G, Valerio MR, Cufari S, Vigneri P, Sanò MV, and Scollo P
- Subjects
- Humans, Female, Retrospective Studies, Middle Aged, Aged, Italy, Adult, Aged, 80 and over, Administration, Oral, Carcinoma, Ovarian Epithelial drug therapy, Treatment Outcome, Neoplasm Recurrence, Local drug therapy, Cyclophosphamide administration & dosage, Cyclophosphamide therapeutic use, Cyclophosphamide adverse effects, Administration, Metronomic, Ovarian Neoplasms drug therapy, Ovarian Neoplasms pathology, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects
- Abstract
Introduction: Metronomic oral cyclophosphamide (MOC) presents many potential advantages, such as significantly less severe side effects than standard regimens, ease of administration, and the delivery of a dose-dense but not necessarily dose-intense treatment. These observations prompted us to evaluate in a retrospective, multicenter study the efficacy and toxicity of MOC in a real-life series of pretreated cancer patients., Methods: The study is a multicenter, retrospective analysis of the activity of single-agent MOC in patients with recurrent or residual epithelial ovarian, fallopian tube, or primary. Eligible patients were continuously treated with MOC at 50 mg/day until progression, toxicity, or death. Overall response rate (ORR), stable disease (SD), and disease control rate (DCR) were reported., Results: The study included 62 patients. Three patients reached a complete response rate (5%), 11 had a partial response rate (18), and 15 had stabilization of disease (24) for an ORR of 23% and a DCR of 47%. Patients with low-grade indolent tumors showed an ORR and an SD rate higher than that observed in non-indolent ones (33% vs. 18% and 28% vs. 14%, respectively). Overall, progression-free survival was 3.5 months (range 1-9 months)., Conclusion: Single-agent MOC is active and very well tolerated in a significant fraction of patients with refractory, recurrent, or residual epithelial ovarian, fallopian tube, or primary peritoneal cancer. In the vision of a practical approach, single-agent MOC may be a useful palliative treatment option for patients with poor tolerance to high-dose regimens or widely pretreated. Further studies are needed better to characterize the role of such an approach in clinical practice., Competing Interests: Declaration of Conflicting InterestsThe author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
- Published
- 2024
- Full Text
- View/download PDF
21. Alkylating agents-induced gonadotoxicity in prepubertal males: Insights on the clinical and preclinical front.
- Author
-
Sriram S, Macedo T, Mavinkurve-Groothuis A, van de Wetering M, and Looijenga LHJ
- Subjects
- Humans, Male, Child, Infertility, Male chemically induced, Infertility, Male etiology, Infertility, Male diagnosis, Animals, Spermatogenesis drug effects, Spermatogenesis radiation effects, Neoplasms drug therapy, Puberty drug effects, Puberty physiology, Alkylating Agents adverse effects, Alkylating Agents administration & dosage, Testis drug effects, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating administration & dosage
- Abstract
Rising cure rates in pediatric cancer patients warrants an increased attention toward the long-term consequences of the diagnosis and treatment in survivors. Chemotherapeutic agents can be gonadotoxic, rendering them at risk for infertility post-survival. While semen cryopreservation is an option that can be provided for most (post)pubertal boys before treatment, this is unfortunately not an option prepubertal in age, simply due to the lack of spermatogenesis. Over the last couple of years, studies have thus focused on better understanding the testis niche in response to various chemotherapeutic agents that are commonly administered and their direct and indirect impact on the germ cell populations. These are generally compounds that have a high risk of infertility and have been classified into risk categories in curated fertility guidelines. However, with it comes the lack of evidence and the challenge of using informative models and conditions most reflective of the physiological scenario, in short, the appropriate study designs for clinically relevant outcomes. Besides, the exact mechanism(s) of action for many of these "risk" compounds as well as other agents is unclear. Understanding their behavior and effect on the testis niche will pave the way for incorporating new strategies to ultimately combat infertility. Of the various drug classes, alkylating agents pose the highest risk of gonadotoxicity as per previously established studies as well as risk stratification guidelines. Therefore, this review will summarize the findings in the field of male fertility concerning gonadotoxicity of akylating agents as a result of chemotherapy exposure., (© 2024 The Author(s). Clinical and Translational Science published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2024
- Full Text
- View/download PDF
22. In Reply: Efficacy and Safety of Carmustine Wafer Implantation After Ventricular Opening in Glioblastomas, Isocitrate Dehydrogenase-Wildtype, in Adults.
- Author
-
Roux A, Elia A, Zanello M, and Pallud J
- Subjects
- Humans, Adult, Glioblastoma drug therapy, Glioblastoma surgery, Carmustine administration & dosage, Isocitrate Dehydrogenase genetics, Brain Neoplasms surgery, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating administration & dosage
- Published
- 2024
- Full Text
- View/download PDF
23. Prolonged 14-day continuous infusion of high-dose ifosfamide for patients with relapsed and refractory high-grade osteosarcoma: a retrospective multicentre cohort study.
- Author
-
Tirtei E, Campello A, Sciannameo V, Asaftei SD, Meazza C, Sironi G, Longhi A, Ibrahim T, Tamburini A, Coccoli L, Crocco F, Cagnazzo C, De Luna E, Quarello P, Berchialla P, and Fagioli F
- Subjects
- Humans, Male, Female, Retrospective Studies, Adult, Adolescent, Young Adult, Middle Aged, Child, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Neoplasm Grading, Treatment Outcome, Ifosfamide administration & dosage, Ifosfamide adverse effects, Ifosfamide therapeutic use, Osteosarcoma drug therapy, Osteosarcoma mortality, Osteosarcoma pathology, Bone Neoplasms drug therapy, Bone Neoplasms mortality, Bone Neoplasms pathology, Neoplasm Recurrence, Local drug therapy
- Abstract
Background: The prognosis of patients with Relapsed/Refractory Osteosarcoma (R/R OS) remains dismal without an agreement on systemic therapy. The use of High-Dose Ifosfamide (14 g/sqm) with an external pump in outpatient setting (14-IFO) in R/R OS patients is limited. This study represents the first retrospective cohort analysis focused on evaluating the activity and toxicity of 14-IFO in this setting., Patients and Methods: The study investigated 14-IFO activity, in terms of tumour response according to RECIST 1.1 criteria, as well as survival rates and toxicity, according to CTCAE v.5., Results: The trial enrolled 26 patients with R/R OS. The Overall Response Rate (ORR) and Disease Control Rate (DCR) obtained was 23% and 57.5%, respectively. Patients with relapsed OS showed a higher ORR (45%) and DCR (82%) compared to refractory patients, irrespective of the number of prior treatment lines received. The achievement of disease control with 14-IFO administration enabled 27% of patients to undergo new local treatment. Four-month Progression-Free Survival (PFS) was 54% for all patients and 82% for the relapsed OS sub-group. Median Overall Survival (OSurv) was 13.7 months, with 1-year OSurv of 51% for all patients and 71% for relapsed patients. Age over 18 years and the presence of refractory disease were identified as negative prognostic factors for this patient cohort. A total of 101 cycles were evaluated for toxic assessment, demonstrating a tolerable profile without grade 3-4 non-haematological toxicities., Conclusions: 14-IFO should be considered a viable treatment option for R/R OS, particularly due to its well tolerated toxicity profile and the potential for home-administration, which can improve patient quality of life without compromising efficacy., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
24. Revisiting intrathecal thiotepa: Efficacy and safety in secondary CNS malignancies.
- Author
-
Jamison T, Haque E, Muhsen IN, Samarkandi H, Fakih RE, and Aljurf M
- Subjects
- Humans, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Agents, Alkylating adverse effects, Hematologic Neoplasms drug therapy, Meningeal Neoplasms drug therapy, Meningeal Neoplasms secondary, Thiotepa administration & dosage, Thiotepa adverse effects, Injections, Spinal, Central Nervous System Neoplasms drug therapy, Central Nervous System Neoplasms secondary
- Abstract
Treating metastatic malignancies to the central nervous system (CNS) is challenging because many drugs cannot cross the blood-brain-barrier (BBB). Direct intrathecal (IT) drug administration into the cerebrospinal fluid (CSF) is a strategy to overcome this problem. Thiotepa has effective CNS penetration but its popularity has waned over the last two decades due to concerns about its efficacy and potential systemic toxicity. This review evaluates the available evidence for the use of IT thiotepa in hematologic malignancies and non-CNS solid tumors with leptomeningeal disease metastases (LMD). Our search shows that IT thiotepa is a reasonable alternative in hematologic malignancies and LMD due to solid organ malignancies. This suggests a potential role of IT thiotepa in second-or third-line treatment or a substitute role in cases of drug-shortages and adverse effects with other agents. Future research should focus on rigorous comparative trials to establish its definitive role in the evolving landscape of CNS-directed chemotherapy., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
25. A case of ifosfamide-induced acute kidney injury, Fanconi syndrome, and nephrogenic diabetes insipidus.
- Author
-
Hoang S, Pujar T, Bellorin-Font E, Edwards JC, and Miyata KN
- Subjects
- Humans, Male, Middle Aged, Deamino Arginine Vasopressin administration & dosage, Deamino Arginine Vasopressin therapeutic use, Hypokalemia chemically induced, Acute Kidney Injury chemically induced, Antineoplastic Agents, Alkylating adverse effects, Diabetes Insipidus, Nephrogenic chemically induced, Fanconi Syndrome chemically induced, Ifosfamide adverse effects
- Abstract
Ifosfamide, a cytotoxic antineoplastic drug, can induce rare complications of Fanconi syndrome and nephrogenic diabetes insipidus (DI). Ifosfamide-induced Fanconi syndrome tends to occur in patients with certain risk factors including young age, high cumulative ifosfamide dose, and coadministration of cisplatin. Nephrogenic DI causes polyuria from impaired urinary concentrating ability due to resistance to arginine vasopressin (AVP) at the collecting duct. These complications are serious and potentially fatal. Here, we describe a case of a middle-aged man without risk factors who was admitted for the management of acute kidney injury and electrolyte derangements after his fourth cycle of chemotherapy including ifosfamide for synovial sarcoma. He was found to have hypokalemia, hypophosphatemia, renal glycosuria, and aminoaciduria, likely from Fanconi syndrome, which were managed by electrolyte replacement therapy. In addition, polyuria and hypernatremia were considered due to nephrogenic DI, which partially responded to desmopressin treatment. This case highlights the importance of the routine electrolytes monitoring after ifosfamide treatment., (© 2023. The Author(s), under exclusive licence to Japanese Society of Nephrology.)
- Published
- 2024
- Full Text
- View/download PDF
26. Treosulfan is a safe and effective alternative to busulfan for conditioning in adult allogeneic HSCT patients: Data from a single center.
- Author
-
Uzay A, Gündoğdu Y, Koşan B, Yetiş T, and Kartı SS
- Subjects
- Humans, Female, Male, Adult, Middle Aged, Retrospective Studies, Young Adult, Adolescent, Treatment Outcome, Aged, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Busulfan analogs & derivatives, Busulfan therapeutic use, Busulfan adverse effects, Busulfan administration & dosage, Transplantation Conditioning methods, Hematopoietic Stem Cell Transplantation adverse effects, Hematopoietic Stem Cell Transplantation methods, Graft vs Host Disease etiology, Graft vs Host Disease prevention & control, Graft vs Host Disease epidemiology, Transplantation, Homologous
- Abstract
Introduction: Type of conditioning regimen impacts the outcome of patients who undergo allogeneic HSCT since graft versus host disease (GVHD), infections, regimen related toxicities (RRT) are important causes of post-transplant mortality. Despite the RRT profile of busulfan, it is frequently used worldwide. Treosulfan has advantages in terms of dose of administration, lower incidence of sinusoidal obstruction syndrome and lower neurotoxicity. We retrospectively investigated outcomes of patients who underwent allogeneic HSCT with treosulfan or busulfan based conditioning regimens in our institution., Methods: Treosulfan was administered to 94 patients while 85 patients received busulfan. Our outcomes were RRT, chronic and acute GVHD, relapse related mortality (RRM), non-relapse mortality, and fungal infection. The clinical follow up data, regarding the primary and secondary endpoints of our study, of the patients who received treosulfan or busulfan based conditioning regimens were statistically analyzed., Results: The median follow-up was 14 months for the treosulfan group while it was 11 months for the busulfan group (p = 0.16). RRT was 11.7% and 7.1% for treosulfan and busulfan respectively. The incidence of extensive chronic GVHD was less frequent in the treosulfan group compared to the busulfan group (15.7% vs. 32.1%) (p < 0.001). The incidence of acute GVHD (Grade 3 or higher) was 32.2% in the treosulfan group while it was 31.6% in the busulfan group. The RRM was 17% in the treosulfan group while it was 34% in the busulfan group. The non-relapse mortality was 35.5% and 29.4% in the treosulfan group and in the busulfan group respectively (p = 0.962)., Conclusion: Treosulfan, with a lower RRM, lower chronic GVHD incidence and with a similar RRT profile appears to be a safe alternative to busulfan., (© 2024 The Author(s). Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
27. Risk factors for nausea and vomiting requiring the daily administration of 5-HT 3 receptor antagonists in radiotherapy combined with temozolomide for high-grade glioma.
- Author
-
Takagi M, Sagara A, Kumakura Y, Watanabe M, Inoue R, Miyazaki M, Ohka F, Motomura K, Natsume A, Wakabayashi T, Saito R, and Yamada K
- Subjects
- Humans, Male, Female, Middle Aged, Risk Factors, Aged, Brain Neoplasms drug therapy, Brain Neoplasms radiotherapy, Adult, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating administration & dosage, Chemoradiotherapy adverse effects, Chemoradiotherapy methods, Temozolomide therapeutic use, Temozolomide administration & dosage, Temozolomide adverse effects, Nausea, Serotonin 5-HT3 Receptor Antagonists therapeutic use, Serotonin 5-HT3 Receptor Antagonists administration & dosage, Vomiting chemically induced, Vomiting drug therapy, Glioma drug therapy, Glioma radiotherapy
- Abstract
Radiotherapy combined with temozolomide (TMZ+RT) is the primary treatment for high-grade glioma. TMZ is classified as a moderate emetic risk agent and, thus, supportive care for nausea and vomiting is important. In Nagoya University Hospital, all patients are treated with a 5-hydroxy-tryptamine 3 receptor antagonist (5-HT
3 RA) for the first 3 days. The daily administration of 5-HT3 RA is resumed after the 4th day based on the condition of patients during TMZ+RT. Therefore, the present study investigated risk factors for nausea and vomiting in patients requiring the daily administration of 5-HT3 RA. Patients with high-grade glioma who received TMZ+RT between January 2014 and December 2019 at our hospital were included. Patients were divided into two groups: a control group (patients who did not resume 5-HT3 RA) and resuming 5-HT3 RA group (patients who resumed 5-HT3 RA after the 4th day), and both groups were compared to identify risk factors for nausea and vomiting during TMZ+RT. There were 78 patients in the control group (68%) and 36 in the resuming 5-HT3 RA group (32%). A multivariate analysis of patient backgrounds in the two groups identified age <18 years, PS 2 or more, and occipital lobe tumors as risk factors for nausea and vomiting. Nausea and vomiting were attenuated in 30 patients (83%) in the resuming 5-HT3 RA group following the resumption of 5-HT3 RA. The results obtained highlight the importance of extracting patients with these risk factors before the initiation of therapy and the early resumption or daily administration of 5-HT3 RA according to the condition of each patient., Competing Interests: The authors declare no conflict of interest.- Published
- 2024
- Full Text
- View/download PDF
28. Single-Agent Trabectedin Versus Physician's Choice Chemotherapy in Patients With Recurrent Ovarian Cancer With BRCA -Mutated and/or BRCAness Phenotype: A Randomized Phase III Trial.
- Author
-
Lorusso D, Raspagliesi F, Ronzulli D, Valabrega G, Colombo N, Pisano C, Cassani C, Tognon G, Tamberi S, Mangili G, Mammoliti S, De Giorgi U, Greco F, Mosconi AM, Breda E, Artioli G, Andreetta C, Casanova C, Ceccherini R, Frassoldati A, Salutari V, Giolitto S, and Scambia G
- Subjects
- Humans, Female, Middle Aged, Aged, Adult, Phenotype, Prospective Studies, BRCA2 Protein genetics, BRCA1 Protein genetics, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Aged, 80 and over, Progression-Free Survival, Trabectedin therapeutic use, Trabectedin administration & dosage, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Ovarian Neoplasms mortality, Mutation, Neoplasm Recurrence, Local drug therapy
- Abstract
Purpose: Literature evidence suggests that trabectedin monotherapy is effective in patients with recurrent ovarian cancer (OC) presenting BRCA mutation and/or BRCAness phenotype., Methods: A prospective, open-label, randomized phase III MITO-23 trial evaluated the activity and safety of trabectedin 1.3 mg/m
2 given once every 3 weeks (arm A) in BRCA 1/2 mutation carriers or patients with BRCAness phenotype (ie, patients who responded to ≥two previous platinum-based treatments) with recurrent OC, primary peritoneal carcinoma, or fallopian tube cancer in comparison with physician's choice chemotherapy in the control arm (arm B; pegylated liposomal doxorubicin, topotecan, gemcitabine, once-weekly paclitaxel, or carboplatin). The primary end point was overall survival (OS) evaluated in the intention-to-treat population., Results: Overall, 244 patients from 21 MITO centers were randomly assigned (arm A = 122/arm B = 122). More than 70% of patients received ≥three previous chemotherapy lines and 35.7% had received a poly (ADP-ribose) polymerase inhibitor (PARPi) before enrollment. Median OS was not significantly different between the arms: arm A: 15.8 versus arm B: 17.9 months ( P = .304). Median progression-free survival was 4.9 months in arm A versus 4.4 months in arm B ( P = .897). Among 208 patients evaluable for efficacy, the objective response rate was 17.1% in arm A and 21.4% in arm B, with comparable median duration of response (5.62 v 5.66 months, respectively). No superior effect was observed for trabectedin in the prespecified subgroup analyses according to BRCA mutational status, chemotherapy type, and pretreatment with a PARPi and/or platinum-free interval. Trabectedin showed a higher frequency of grade ≥3 adverse events (AEs), serious AEs, and serious adverse drug reactions compared with control chemotherapy., Conclusion: Trabectedin did not improve median OS and showed a worse safety profile in comparison with physician's choice control chemotherapy.- Published
- 2024
- Full Text
- View/download PDF
29. Association of temozolomide with progressive multifocal leukoencephalopathy: a disproportionality analysis integrated with network pharmacology.
- Author
-
Bhati V, Kumar A, Lather V, Sharma R, and Pandita D
- Subjects
- Humans, Female, Male, Middle Aged, Adult, United States, Aged, Glioblastoma drug therapy, Molecular Docking Simulation, United States Food and Drug Administration, Sex Factors, Temozolomide administration & dosage, Temozolomide pharmacology, Temozolomide adverse effects, Leukoencephalopathy, Progressive Multifocal chemically induced, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating administration & dosage, Adverse Drug Reaction Reporting Systems statistics & numerical data, Databases, Factual, Network Pharmacology
- Abstract
Background: Temozolomide (TMZ) is an alkylating agent approved for the management of glioblastoma. The TMZ is not known for progressive multifocal leukoencephalopathy (PML). The main objective of the current study is to find out the association of TMZ with PML using disproportionality analysis of FDA Adverse Event Reporting System (FAERS) data integrated with network pharmacological approaches., Research Design and Methods: OpenVigil tool was used to query the FAERS database. The disproportionality measures were calculated. The network has been constructed using Cytoscape. Finally, the possible binding interactions were studied using Glide, Schrödinger Inc., Results: A total number of 3502 cases of PML were reported in the FAERS database. Out of these, 10 cases were found with TMZ. The subgroup analysis results have shown a greater number of cases in females. The network has indicated the involvement of human mitogen-activated protein kinase, 3-phosphoinositide-dependent protein kinase 1 protein, human mTOR complex protein, phosphatidylinositol 4,5-bisphosphate 3-kinase protein, and glycogen synthase kinase-3 beta protein. The docking results have indicated good interactions of TMZ with active site of glycogen synthase kinase-3 beta and mitogen-activated protein kinase 1 as compared to other identified targets., Conclusion: The PML is identified as novel signal with temozolomide.
- Published
- 2024
- Full Text
- View/download PDF
30. Noni enhances the anticancer activity of cyclophosphamide and suppresses myelotoxicity and hepatotoxicity in tumor-bearing mice.
- Author
-
Ali M, Manjula SN, Mohiuddin I, Mruthunjaya K, Shakeel F, Mir SA, and Wani SUD
- Subjects
- Animals, Mice, Humans, Female, Cell Line, Tumor, Fruit and Vegetable Juices, Xenograft Model Antitumor Assays, Drug Synergism, Plant Extracts pharmacology, Antineoplastic Combined Chemotherapy Protocols pharmacology, Antineoplastic Agents, Alkylating pharmacology, Antineoplastic Agents, Alkylating adverse effects, Mice, Inbred BALB C, Chemical and Drug Induced Liver Injury prevention & control, Chemical and Drug Induced Liver Injury etiology, Cyclophosphamide pharmacology, Cyclophosphamide adverse effects, Morinda chemistry, Breast Neoplasms drug therapy, Breast Neoplasms pathology
- Abstract
Background and Aim: Morinda citrifolia fruit juice (noni) is an herbal remedy documented to have antioxidant properties. It has been suggested that prevention of carcinogen-DNA adduct formation and the antioxidant activity of NJ may contribute to the cancer preventive effect. In the present study, the antitumor activity of noni was investigated in the presence of cyclophosphamide (CYL) in vitro and in vivo., Methods: In vitro breast cancer cells (MDA-MB-468) were used to measure the percentage of inhibition and the IC
50 . The in vivo antitumor activity of noni was studied by monitoring the mean survival time (MST), percentage increase in life span (%ILS), viable and non-viable cell count, tumor volume, body weight, and hematological and serum biochemical parameters in mice. Treatment with noni and CYL exhibited dose- and time-dependent cytotoxicity toward breast cancer cells., Results: Individual treatment of noni and CYL exhibited dose- and time-dependent cytotoxicity on breast cancer cell lines, while in combination therapy of noni and CYL, noni enhances cytotoxic effect of CYL at 48 h than that at 24 h. Similar result was found in in vivo studies, the results of which revealed that alone treatment of CYL and noni suppressed tumor growth. However, combination treatment with CYL and noni presented better tumor inhibition than that of alone treatment of CYL and noni. On the contrary, CYL alone drastically attenuated hematological parameters, i.e., RBC, WBC, and Hb compared to normal and control groups, and this change was reversed and normalized by noni when given as combination therapy with CYL. Moreover, the levels of serum biochemical markers, i.e., AST, ALP, and ALT, were significantly increased in the control and CYL-treated groups than those in the normal group. In the combination treatment of noni and CYL, the above biochemical marker levels significantly decreased compared to CYL alone-treated group., Conclusions: The present study suggested that CYL treatment can cause serious myelotoxicity and hepatic injury in cancer patients. In conclusion, the combined use of noni with CYL potentially enhances the antitumor activity of CYL and suppresses myelotoxicity and hepatotoxicity induced by CYL in tumor-bearing mice., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
31. Efficacy and safety of bendamustine for lymphodepletion before lisocabtagene maraleucel.
- Author
-
Ghilardi G, Paruzzo L, Patel V, Svoboda J, Chong ER, Fardella E, Chong EA, Gabrielli G, Nasta SD, Landsburg DJ, Carter J, Pajarillo R, Barta SK, White G, Weber E, Napier E, Porter DL, Garfall AL, Schuster SJ, and Ruella M
- Subjects
- Humans, Middle Aged, Male, Female, Aged, Retrospective Studies, Adult, Lymphoma, Large B-Cell, Diffuse drug therapy, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Biological Products therapeutic use, Biological Products adverse effects, Aged, 80 and over, Treatment Outcome, Bendamustine Hydrochloride therapeutic use, Immunotherapy, Adoptive methods, Immunotherapy, Adoptive adverse effects
- Abstract
Bendamustine has been retrospectively shown to be an effective and safe lymphodepletion regimen prior to the anti-CD19 chimeric antigen receptor T cell (CART) products tisagenlecleucel and axicabtagene ciloleucel, as well as the anti-BCMA CART products idecabtagene vicleucel and ciltacabtagene autoleucel. However, bendamustine as lymphodepletion prior to lisocabtagene maraleucel (liso-cel), a 4-1BB co-stimulated, fixed CD4:CD8 ratio anti-CD19 CART product, has not been described yet. Thus, we studied a cohort of sequentially-treated patients with large B-cell lymphomas who received bendamustine lymphodepletion before liso-cel at the University of Pennsylvania between 5/2021 and 12/2023 (n = 31). Patients were evaluated for toxicities and responses. Of note, 7 patients (22.6%) would have dnot met the inclusion criteria for the registrational liso-cel clinical trials, mostly due to older age. Overall and complete response rates were 76.9% and 73.1%, respectively. At a median follow-up of 6.3 months, the 6-month progression-free and overall survival were 59.9% and 91.1%, respectively. Rates of cytokine-release syndrome (CRS) and neurotoxicity (ICANS) of any grade were 9.7% and 9.7%, respectively, with no grade ≥ 3 events. No infections were reported during the first 30 days following liso-cel infusion. Neutropenia ≥ grade 3 was observed in 29.0% of patients; thrombocytopenia ≥ grade 3 occurred in 9.7%. In conclusion, bendamustine lymphodepletion before liso-cel appears to be a strategy that can drive tumor responses while ensuring a mild toxicity profile., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
32. A Review of Approaches to Potentiate the Activity of Temozolomide against Glioblastoma to Overcome Resistance.
- Author
-
Karve AS, Desai JM, Gadgil SN, Dave N, Wise-Draper TM, Gudelsky GA, Phoenix TN, DasGupta B, Yogendran L, Sengupta S, Plas DR, and Desai PB
- Subjects
- Humans, Temozolomide pharmacology, Temozolomide therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Quality of Life, Neoplasm Recurrence, Local drug therapy, DNA pharmacology, Drug Resistance, Neoplasm genetics, Cell Line, Tumor, Glioblastoma metabolism, Brain Neoplasms pathology
- Abstract
A glioblastoma (GBM) is one of the most aggressive, infiltrative, and treatment-resistant malignancies of the central nervous system (CNS). The current standard of care for GBMs include maximally safe tumor resection, followed by concurrent adjuvant radiation treatment and chemotherapy with the DNA alkylating agent temozolomide (TMZ), which was approved by the FDA in 2005 based on a marginal increase (~2 months) in overall survival (OS) levels. This treatment approach, while initially successful in containing and treating GBM, almost invariably fails to prevent tumor recurrence. In addition to the limited therapeutic benefit, TMZ also causes debilitating adverse events (AEs) that significantly impact the quality of life of GBM patients. Some of the most common AEs include hematologic (e.g., thrombocytopenia, neutropenia, anemia) and non-hematologic (e.g., nausea, vomiting, constipation, dizziness) toxicities. Recurrent GBMs are often resistant to TMZ and other DNA-damaging agents. Thus, there is an urgent need to devise strategies to potentiate TMZ activity, to overcome drug resistance, and to reduce dose-dependent AEs. Here, we analyze major mechanisms of the TMZ resistance-mediated intracellular signaling activation of DNA repair pathways and the overexpression of drug transporters. We review some of the approaches investigated to counteract these mechanisms of resistance to TMZ, including the use of chemosensitizers and drug delivery strategies to enhance tumoral drug exposure.
- Published
- 2024
- Full Text
- View/download PDF
33. A combined analysis of two prospective randomised studies exploring the impact of extended post-radiation temozolomide on survival outcomes in newly diagnosed glioblastoma.
- Author
-
Gately L, Mesía C, Sepúlveda JM, Del Barco S, Pineda E, Gironés R, Fuster J, Hong W, Dumas M, Gill S, Navarro LM, Herrero A, Dowling A, de Las Peñas R, Vaz MA, Alonso M, Lwin Z, Harrup R, Peralta S, Long A, Perez-Segura P, Ahern E, Garate CO, Wong M, Campbell R, Cuff K, Jennens R, Gallego O, Underhill C, Martinez-Garcia M, Covela M, Cooper A, Brown S, Rosenthal M, Torres J, Collins IM, Gibbs P, and Balana C
- Subjects
- Humans, Temozolomide therapeutic use, Prospective Studies, Dacarbazine adverse effects, Disease-Free Survival, Antineoplastic Agents, Alkylating adverse effects, Glioblastoma drug therapy, Glioblastoma radiotherapy, Brain Neoplasms drug therapy, Brain Neoplasms radiotherapy
- Abstract
Purpose: The optimal duration of post-radiation temozolomide in newly diagnosed glioblastoma remains unclear, with no published phase III randomised trials. Standard-of-care stipulates 6 months. However, in routine care, it is often extended to 12 months, despite lacking robust supporting data., Methods: GEINO14-01 (Spain) and EX-TEM (Australia) studies enrolled glioblastoma patients without progression at the end of 6 months post-radiation temozolomide. Participants were randomised 1:1 to six additional months of temozolomide or observation. Primary endpoint was 6-month progression free survival from date of randomisation (6mPFS). Secondary endpoints included overall survival (OS) and toxicity. 204 patients were required to detect an improvement in 6mPFS from 50 to 60% (80% power). Neither study recruited sufficient patients. We performed a combined analysis of individual patient data., Results: 205 patients were recruited: 159 in GEINO14-01 (2014-2018) and 46 in EX-TEM (2019-2022). Median follow-up was 20.0 and 14.5 months. Baseline characteristics were balanced. There was no significant improvement in 6mPFS (57.2% vs 64.0%, OR0.75, p = 0.4), nor across any subgroups, including MGMT methylated; PFS (HR0.92, p = 0.59, median 7.8 vs 9.7 months); or OS (HR1.03, p = 0.87, median 20.1 vs 19.4 months). During treatment extension, 64% experienced any grade adverse event, mainly fatigue and gastrointestinal (both 54%). Only a minority required treatment changes: 4.5% dose delay, 7.5% dose reduction, 1.5% temozolomide discontinuation., Conclusion: For glioblastoma patients, extending post-radiation temozolomide from 6 to 12 months is well tolerated but does not improve 6mPFS. We could not identify any subset that benefitted from extended treatment. Six months should remain standard-of-care., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
34. Ifosfamide-induced nephrotoxicity in oncological patients.
- Author
-
Quiroz-Aldave JE, Durand-Vásquez MDC, Chávez-Vásquez FS, Rodríguez-Angulo AN, Gonzáles-Saldaña SE, Alcalde-Loyola CC, Coronado-Arroyo JC, Zavaleta-Gutiérrez FE, Concepción-Urteaga LA, Haro-Varas JC, and Concepción-Zavaleta MJ
- Subjects
- Humans, Kidney, Ifosfamide adverse effects, Antineoplastic Agents, Alkylating adverse effects
- Abstract
Introduction: Ifosfamide is an alkylating chemotherapeutic agent used in the treatment of various neoplasms. Its main adverse effects include renal damage., Areas Covered: A comprehensive review was conducted, including 100 articles from the Scielo, Scopus, and EMBASE databases. Ifosfamide-induced nephrotoxicity is attributed to its toxic metabolites, such as acrolein and chloroacetaldehyde, which cause mitochondrial damage and oxidative stress in renal tubular cells. Literature review found a 29-year average age with no gender predominance and a mortality of 13%. Currently, no fully effective strategy exists for preventing ifosfamide-induced nephrotoxicity; however, hydration, forced diuresis, and other interventions are employed to limit renal damage. Long-term renal function monitoring is essential for patients treated with ifosfamide., Expert Opinion: Ifosfamide remains essential in neoplasm treatment, but nephrotoxicity, often compounded by coadministered drugs, poses diagnostic challenges. Preventive strategies are lacking, necessitating further research. Identifying timely risk factors can mitigate renal damage, and a multidisciplinary approach manages established nephrotoxicity. Emerging therapies may reduce ifosfamide induced nephrotoxicity.
- Published
- 2024
- Full Text
- View/download PDF
35. Neuropsychological functioning during chemotherapy with temozolomide in high-grade glioma patients: a retrospective single centre study.
- Author
-
Abete-Fornara G, Bintintan Socaciu P, Fanizzi C, Fiore G, Locatelli M, and Caroli M
- Subjects
- Adult, Humans, Temozolomide therapeutic use, Retrospective Studies, Dacarbazine adverse effects, Neoplasm Recurrence, Local drug therapy, Antineoplastic Agents, Alkylating adverse effects, Brain Neoplasms pathology, Glioma pathology
- Abstract
Purpose: starting from a lack of precise and coherent data in literature, aim of this work is to retrospectively study the influence of chemotherapy with Temozolomide (TMZ) on a wide series of neuropsychological functions in a population of adult high-grade glioma patients., Methods: an extensive neuropsychological battery was administered pre-operatively (T0) and after 6 (T1) and 12 months (T2) from surgery. After full recovery from surgery, TMZ was delivered concomitant to radiotherapy and, subsequently, adjuvantly for 5-day cycles per month. Parametric and non-parametric analyses were conducted to verify the influence of several aspects of chemotherapy on the adjusted scores of each cognitive test at the two post-operative follow-ups., Results: Sixty-one patients were included at T0; patients with a lower adjuvant TMZ dosage reported a better performance at the visual attention test at T1, and at the deductive reasoning test at T2. Undergoing more than 8 cycles of adjuvant therapy was slightly associated with a better performance at the long-term verbal memory tasks at T2. No other associations were found with the other cognitive tests and autonomy scales administered., Conclusions: TMZ proved to be a secure treatment with no negative side effects on cognition and on level of daily autonomy, even at the highest dosage used. This is a positive finding which enables clinicians to reassure patients about the absence of significant negative effects of TMZ on their daily life functioning. In this view, eventual cognitive changes during treatment might not be attributed to chemotherapy but to other events such as tumour relapse., (© 2023. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
36. Treatment of glioblastoma in Greenlandic patients.
- Author
-
Frandsen S, Pedersen AJ, Gredal O, Møller S, Geissler UW, and Nørøxe DS
- Subjects
- Adult, Humans, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Dacarbazine therapeutic use, Dacarbazine adverse effects, Chemotherapy, Adjuvant, Temozolomide therapeutic use, Glioblastoma drug therapy, Glioblastoma pathology, Brain Neoplasms drug therapy, Brain Neoplasms pathology
- Abstract
Glioblastoma (GBM), WHO grade IV, is the most common primary malignant brain tumour among adults with a devastating overall survival of 14-22 months. Standard treatment of GBM includes maximum safe resection, radiotherapy plus concomitant and adjuvant temozolomide (TMZ), given over a period of approximately 9 months. Treatment and follow-up for Greenlandic patients with GBM are managed at Rigshospitalet (RH), Copenhagen. Greenlandic GBM patients, therefore, travel back and forth to RH, often unaccompanied, and challenged by cognitive failure or other symptoms from their disease and/or treatment. Few Greenlandic patients are diagnosed with GBM annually, but considering the poor prognosis and short remaining lifespan, it would be preferable to limit their travels. TMZ is administrated as capsules. Health personnel at Queen Ingrid's Hospital (DIH), Nuuk, are trained in treating other oncological diseases and handling side effects. Hence, it could be investigated whether administration of adjuvant TMZ at DIH could be feasible after personnel education as well as economic consideration and compensation, in close collaboration with neuro oncologists at RH. In this article, we describe the Greenlandic cancer treatment, and the typical workflow from diagnosis of GBM to treatment to progression.
- Published
- 2023
- Full Text
- View/download PDF
37. Phase 1 dose-escalation study evaluating the safety, pharmacokinetics, and clinical activity of OBI-3424 in patients with advanced or metastatic solid tumors.
- Author
-
Tsimberidou AM, Verschraegen CF, Wesolowski R, Shia CS, Hsu P, and Pearce TE
- Subjects
- Humans, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating pharmacology, Dose-Response Relationship, Drug, Anemia chemically induced, Anemia drug therapy, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacology, Neoplasms drug therapy, Neoplasms pathology, Neoplasms, Second Primary, Thrombocytopenia chemically induced
- Abstract
Background: Report of a Phase 1 dose-escalation study of OBI-3424 monotherapy in patients with advanced solid tumors (NCT03592264)., Methods: A classic 3 + 3 design was used to determine the maximum tolerated dose and recommended Phase 2 dose (RP2D) of OBI-3424 administered intravenously, as a single agent, at doses of 1, 2, 4, 6, 8, or 12 mg/m
2 (days 1 and 8 of a 21-day cycle, Schedule A) or 8, 10, 12, or 14 mg/m2 (day 1 of a 21-day cycle, Schedule B)., Results: Dose-limiting hematologic toxicities at 12 mg/m2 in Schedule A led to dose and schedule modifications (Schedule B). In Schedule B, maximum tolerated dose was not reached at the maximum dose tested (14 mg/m2 ). Grade ≥3 anemia was noted in 3/6 patients treated at 14 mg/m2 ; the RP2D was 12 mg/m2 (Schedule B). Grade ≥3 treatment-emergent adverse events were experienced by 19/39 (49%) and included anemia (41%) and thrombocytopenia (26%); three patients experienced serious treatment-emergent adverse events (grade ≥3 anemia and thrombocytopenia). One patient had a partial response and 21/33 (64%) had stable disease., Conclusions: The RP2D is 12 mg/m2 once every 3 weeks. OBI-3424 was well tolerated; dose-dependent, noncumulative thrombocytopenia and anemia were dose-limiting., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
38. Toxicity to intravitreal melphalan in a patient with retinoblastoma.
- Author
-
Solaz-Ruiz MG, Pérez LA, Cauto-Picazo C, Barranco-González H, Pascual-Camps I, and España-Gregori E
- Subjects
- Humans, Melphalan adverse effects, Antineoplastic Agents, Alkylating adverse effects, Retrospective Studies, Vitreous Body, Intravitreal Injections, Neoplasm Seeding, Retinoblastoma diagnosis, Retinoblastoma drug therapy, Retinal Neoplasms diagnosis, Retinal Neoplasms drug therapy
- Abstract
Objective: Description of melphalan's toxicity in retinoblastoma treatment. Methods: Clinical case report. Results: We presented a case of unilateral retinoblastoma with vitreous seeding at diagnosis, in which the use of intravitreal melphalan produced many adverse reactions. Conclusions: Vitreous seedings have been one of the most important challenges in retinoblastoma treatment. Intravitreal melphalan has achieved the regression of vitreous seedings in a large percentage of cases. It is a safe treatment; however, it can produce toxicity, even with the standard dose of 20-30 µg, which has been poorly documented. Exhaustive follow-up of patients is recommended for an early diagnosis of possible adverse effects. Abbreviations: OS = left eye, RI = magnetic resonance imaging, OCT = optical coherence tomography., (#x00A9; The Authors.Romanian Society of Ophthalmology.)
- Published
- 2023
- Full Text
- View/download PDF
39. Comparative study of concurrent conventional chemoradiotherapy versus hypofractionated chemoradiotherapy in newly diagnosed glioblastoma multiforme postoperative patients.
- Author
-
Khatri NK, Kumar HS, Sharma N, Jakhar SL, and Dhaka S
- Subjects
- Humans, Antineoplastic Agents, Alkylating adverse effects, Chemoradiotherapy adverse effects, Temozolomide adverse effects, Brain Neoplasms therapy, Brain Neoplasms drug therapy, Glioblastoma therapy, Glioblastoma drug therapy
- Abstract
Purpose: To assess the treatment response and toxicity profile among two groups of newly diagnosed glioblastoma multiforme (GBM) postoperative patients receiving conventional radiotherapy (RT) versus hypofractionated RT with concurrent temozolomide (TMZ) in both., Materials and Methods: A total of 50 patients randomly allotted into two arms (25 in each). Dose received 60 Gy (2 Gy/#) in conventional fractionation RT versus 50 Gy (2.5 Gy/#) in hypofractionated RT with concurrent TMZ 75 mg/m
2 orally daily in both arms, respectively. Follow-up was done at 1, 3, 6, and 12 months after completion of treatment to evaluate toxicities, treatment response, and progression-free survival (PFS)., Results: All patients were well tolerated with treatment; no major adverse effects were monitored in two arms. There was no statistical significant difference in treatment response, which was found 64% versus 60% in arm A and arm B, respectively, at 3 months of follow-up (P = 0.768). Toxicity profiles were also noted similar in both arms. The 6-month PFS was 84% and 80% in arm A and arm B, respectively (P = 0.71) and 12-month PFS was 60% and 52% in arm A and arm B, respectively (P = 0.69)., Conclusion: Among the patients followed, this study showed that hypofractionated RT regimen was not inferior to conventional RT regimen.- Published
- 2023
- Full Text
- View/download PDF
40. Safe administration of temozolomide in end-stage renal disease patients.
- Author
-
Hundal J, Singh A, Pereira MK, and Vredenburg J
- Subjects
- Male, Humans, Middle Aged, Temozolomide therapeutic use, Renal Dialysis, Antineoplastic Agents, Alkylating adverse effects, Glioblastoma pathology, Kidney Failure, Chronic therapy, Central Nervous System Neoplasms drug therapy, Brain Neoplasms drug therapy
- Abstract
Introduction: Glioblastoma multiforme is the most common and aggressive type of central nervous system tumor. We present a novel case of the challenges of dosing temozolomide in a patient with end stage renal disease on peritoneal hemodialysis with unpredictable clearance and toxicities., Case Report: In this case, a 60-year-old male with a past medical history of hypertension and Stage V chronic kidney disease presented with worsening confusion and word-finding difficulty in the emergency department. Magnetic resonance imaging demonstrated a large intra-axial mass within the posterior left frontal lobe measuring 4.5 × 4.1 × 3.5 cm with irregularly, predominant peripheral tumoral enhancement., Management and Outcome: The patient underwent a surgical resection which confirmed the diagnosis of glioblastoma (grade 4). The standard treatment for glioblastoma is 6 weeks of radiation therapy and daily temozolomide. Given his history of renal dysfunction and limited data on the safety of temozolomide in patients on hemodialysis (HD), the patient was administered dose-reduced temozolomide and closely monitored for toxicities. Temozolomide was successfully up-titrated to the full dose., Discussion: Renal replacement therapy is a life-saving treatment for end-stage kidney disease patients. A stepwise increase in the dosage of temozolomide did not increase the risk of toxicity with HD. There are no studies with patients on temozolomide and peritoneal dialysis. Our case transitioned to peritoneal dialysis from HD without significant toxicity from temozolomide. As a more substantial proportion of the population becomes dialysis-dependent in the coming years, we need further studies to understand the safety profiles of chemotherapeutic agents in this complex subset of patients.
- Published
- 2023
- Full Text
- View/download PDF
41. Effectiveness and Safety of Trabectedin and Radiotherapy for Patients With Myxoid Liposarcoma: A Nonrandomized Clinical Trial.
- Author
-
Sanfilippo R, Hindi N, Cruz Jurado J, Blay JY, Lopez-Pousa A, Italiano A, Alvarez R, Gutierrez A, Rincón-Perez I, Sangalli C, Pérez Aguiar JL, Romero J, Morosi C, Sunyach MP, Fabbroni C, Romagosa C, Ranchere-Vince D, Dei Tos AP, Casali PG, Martin-Broto J, and Gronchi A
- Subjects
- Adult, Humans, Male, Female, Trabectedin adverse effects, Trabectedin administration & dosage, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating administration & dosage, Neoplasm Recurrence, Local drug therapy, Liposarcoma, Myxoid drug therapy, Liposarcoma, Myxoid radiotherapy, Sarcoma drug therapy, Soft Tissue Neoplasms drug therapy
- Abstract
Importance: Preclinical data about the synergistic activity of radiotherapy (RT) and trabectedin have been reported. The combination of trabectedin and RT in treating myxoid liposarcomas appears worth exploring., Objective: To explore the effectiveness and safety of trabectedin combined with RT., Design, Setting, and Participants: This international, open-label, phase 2 nonrandomized clinical trial including 46 patients with myxoid liposarcoma was conducted in 4 centers in Spain, 1 in Italy, and 2 in France from July 1, 2016, to September 30, 2019. Eligible patients had to have a histologic, centrally reviewed diagnosis of localized resectable myxoid liposarcoma arising from an extremity or the trunk wall., Interventions: Trabectedin was administered at the recommended dose stemming from the phase 1 trial (1.5 mg/m2), with intravenous infusion during 24 hours every 21 days for a total of 3 cycles. Radiotherapy was started after completion of the first trabectedin infusion (cycle 1, day 2). Patients received 25 fractions of radiation for a total of 45 Gy. Surgery was planned 3 to 4 weeks after the administration of the last preoperative cycle and not until 4 weeks after the end of preoperative RT. Pathologic specimens were mapped in tumor sections to estimate the histologic changes and the percentage of viable tumor after neoadjuvant treatment., Main Outcomes and Measures: The primary objective of the phase 2 part of the study was overall response. Secondary objectives were effectiveness measured by relapse-free survival and activity measured by functional imaging and pathologic response., Results: A total of 46 patients were enrolled. Four patients were not evaluable. The median age was 43 years (range, 18-77 years), and 31 patients were male (67%). Overall, 9 of 41 patients (22%) achieved a partial response with neoadjuvant treatment with trabectedin and RT, with 5 of 39 patients (13%) achieving a complete pathologic response and 20 of 39 patients (51%) having 10% or less of a viable remaining tumor. Partial responses according to Choi criteria were observed in 24 of 29 evaluable patients (83%), and no patient had disease progression. Treatment was well tolerated., Conclusions and Relevance: Although the primary end point of this phase 2 nonrandomized clinical trial was not met (Response Evaluation Criteria in Solid Tumors response in ≥70% of patients), results suggest this combination was well tolerated and effective in terms of pathologic response. Thus, trabectedin plus RT might be a treatment option regarding tolerability; further evidence should be generated in this setting.
- Published
- 2023
- Full Text
- View/download PDF
42. Exploring temozolomide encapsulated PEGylated liposomes and lyotropic liquid crystals for effective treatment of glioblastoma: in-vitro, cell line, and pharmacokinetic studies.
- Author
-
Waghule T, Laxmi Swetha K, Roy A, Narayan Saha R, and Singhvi G
- Subjects
- Liquid Crystals, Polyethylene Glycols, Humans, Half-Life, Glioblastoma drug therapy, Brain Neoplasms drug therapy, Tissue Distribution, Blood-Brain Barrier metabolism, Nanoparticle Drug Delivery System, Antineoplastic Agents, Alkylating administration & dosage, Antineoplastic Agents, Alkylating adverse effects, Antineoplastic Agents, Alkylating pharmacokinetics, Male, Animals, Rats, Temozolomide administration & dosage, Temozolomide adverse effects, Temozolomide pharmacokinetics, Liposomes
- Abstract
Temozolomide (TMZ) is one of the best choices for treating glioblastoma. However, due to the short plasma half-life, only 20-30 % brain bioavailability can be achieved using traditional formulations. In the present study, PEGylated liposomes and lyotropic liquid crystals (LLCs) were developed and investigated to prolong the plasma circulation time of TMZ. Industrially feasible membrane extrusion and modified hot melt emulsification techniques were utilized during the formulation. Liposomes and LLCs in the particle size range of 80-120 nm were obtained with up to 50 % entrapment efficiency. The nanocarriers were found to show a prolonged release of up to 72 h. The cytotoxicity studies in glioblastoma cell lines revealed a ∼1.6-fold increased cytotoxicity compared to free TMZ. PEGylated liposomes and PEGylated LLCs were found to show a 3.47 and 3.18-fold less cell uptake in macrophage cell lines than uncoated liposomes and LLCs, respectively. A 1.25 and 2-fold increase in the plasma t
1/2 was observed with PEGylated liposomes and PEGylated LLCs, respectively, compared to the TMZ when administered intravenously. Extending plasma circulation time of TMZ led to significant increase in brain bioavailability. Overall, the observed improved pharmacokinetics and biodistribution of TMZ revealed the potential of these PEGylated nanocarriers in the efficient treatment of glioblastoma., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier B.V. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
43. Addition of intravitreal carboplatin with melphalan for management of vitreous seeding in retinoblastoma.
- Author
-
Riazi-Esfahani H, Masoomian B, Khodabandeh A, Amini A, Taghizadeh S, Boujabadi L, Sharifkashani S, Shields CL, and Ghassemi F
- Subjects
- Humans, Infant, Melphalan adverse effects, Carboplatin therapeutic use, Retrospective Studies, Antineoplastic Agents, Alkylating adverse effects, Neoplasm Recurrence, Local pathology, Vitreous Body pathology, Neoplasm Seeding, Intravitreal Injections, Retinoblastoma diagnosis, Retinoblastoma drug therapy, Retinal Neoplasms diagnosis, Retinal Neoplasms drug therapy
- Abstract
Purpose: To evaluate the efficacy and toxicity of intravitreal carboplatin plus melphalan for the treatment of vitreous seeds in eyes with retinoblastoma (RB)., Methods: This retrospective series at a tertiary referral center included 22 consecutive RB patients who had received intravitreal carboplatin (16 μg per 0.05 ml) combined with melphalan (30 μg in 0.03 ml) [IVi (Ca-Me)] for treatment of vitreous seeds. Tumor control and drug toxicities were recorded., Results: There were 22 eyes of 22 patients, divided into primary group (n = 13) without history of previous intravitreal chemotherapy (IViC) and refractory group (n = 9) with history of previous IViC using melphalan and/or topotecan. The demographics and clinical findings of the primary and refractory groups did not differ significantly. The 6-month follow-up revealed complete vitreous seed control (77% vs. 89%, p = 0.47). Vitreous seed recurrence was detected in 1 eye of each group at 6 months. During the next 18-month follow-up period, no recurrence of seed was observed. The response to IVi (Ca-Me) was not significantly influenced by previous IViC (p = 0.70), primary systemic or intra-arterial chemotherapy (p = 0.45), or the type of regression (p = 0.35). The most common tumor treatment complications were retinal detachment (RD) (n = 2), early hypotony (n = 2) and late hypotony (n = 4, unrelated), cataract (n = 2), and severe pigment dispersion (n = 1). Enucleation was performed in 8 eyes, for total RD (n = 1), phthisis bulbi (n = 5), and extensive solid tumor recurrence (n = 2). There was no case of orbital invasion, systemic metastasis, or death., Conclusion: Based on this interventional case series for primary and refractory vitreous RB seeds, carboplatin plus melphalan therapy may be effective with few toxic side effects., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
44. Letter to the editor regarding "The efficacy and safety of radiotherapy with adjuvant temozolomide for glioblastoma: A meta-analysis of randomized controlled studies".
- Author
-
Yang L, Ye Z, and Huang Q
- Subjects
- Humans, Antineoplastic Agents, Alkylating adverse effects, Chemotherapy, Adjuvant, Dacarbazine therapeutic use, Radiotherapy, Adjuvant, Temozolomide therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms radiotherapy, Glioblastoma drug therapy, Glioblastoma radiotherapy
- Abstract
Competing Interests: Competing interests The authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
45. Adjuvant Temozolomide Chemotherapy With or Without Interferon Alfa Among Patients With Newly Diagnosed High-grade Gliomas: A Randomized Clinical Trial.
- Author
-
Guo C, Yang Q, Xu P, Deng M, Jiang T, Cai L, Li J, Sai K, Xi S, Ouyang H, Liu M, Li X, Li Z, Ni X, Cao X, Li C, Wu S, Du X, Su J, Xue X, Wang Y, Li G, Qin Z, Yang H, Zhou T, Liu J, Hu X, Wang J, Jiang X, Lin F, Zhang X, Ke C, Lv X, Lv Y, Hu W, Zeng J, Chen Z, Zhong S, Wang H, Chen Y, Zhang J, Li D, Mou Y, and Chen Z
- Subjects
- Female, Humans, Male, Middle Aged, Antineoplastic Agents, Alkylating therapeutic use, Antineoplastic Agents, Alkylating adverse effects, Dacarbazine therapeutic use, Interferon-alpha therapeutic use, Temozolomide therapeutic use, Adolescent, Young Adult, Adult, Aged, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Glioma drug therapy
- Abstract
Importance: High-grade gliomas (HGGs) constitute the most common and aggressive primary brain tumor, with 5-year survival rates of 30.9% for grade 3 gliomas and 6.6% for grade 4 gliomas. The add-on efficacy of interferon alfa is unclear for the treatment of HGG., Objectives: To compare the therapeutic efficacy and toxic effects of the combination of temozolomide and interferon alfa and temozolomide alone in patients with newly diagnosed HGG., Design, Setting, and Participants: This multicenter, randomized, phase 3 clinical trial enrolled 199 patients with newly diagnosed HGG from May 1, 2012, to March 30, 2016, at 15 Chinese medical centers. Follow-up was completed July 31, 2021, and data were analyzed from September 13 to November 24, 2021. Eligible patients were aged 18 to 75 years with newly diagnosed and histologically confirmed HGG and had received no prior chemotherapy, radiotherapy, or immunotherapy for their HGG., Interventions: All patients received standard radiotherapy concurrent with temozolomide. After a 4-week break, patients in the temozolomide with interferon alfa group received standard temozolomide combined with interferon alfa every 28 days. Patients in the temozolomide group received standard temozolomide., Main Outcomes and Measures: The primary end point was 2-year overall survival (OS). Secondary end points were 2-year progression-free survival (PFS) and treatment tolerability., Results: A total of 199 patients with HGG were enrolled, with a median follow-up time of 66.0 (95% CI, 59.1-72.9) months. Seventy-nine patients (39.7%) were women and 120 (60.3%) were men, with ages ranging from 18 to 75 years and a median age of 46.9 (95% CI, 45.3-48.7) years. The median OS of patients in the temozolomide plus interferon alfa group (26.7 [95% CI, 21.6-31.7] months) was significantly longer than that in the standard group (18.8 [95% CI, 16.9-20.7] months; hazard ratio [HR], 0.64 [95% CI, 0.47-0.88]; P = .005). Temozolomide plus interferon alfa also significantly improved median OS in patients with O6-methylguanine-DNA methyltransferase (MGMT) unmethylation (24.7 [95% CI, 20.5-28.8] months) compared with temozolomide (17.4 [95% CI, 14.1-20.7] months; HR, 0.57 [95% CI, 0.37-0.87]; P = .008). Seizure and influenzalike symptoms were more common in the temozolomide plus interferon alfa group, with 2 of 100 (2.0%) and 5 of 100 (5.0%) patients with grades 1 and 2 toxic effects, respectively (P = .02). Finally, results suggested that methylation level at the IFNAR1/2 promoter was a marker of sensitivity to temozolomide plus interferon alfa., Conclusions and Relevance: Compared with the standard regimen, temozolomide plus interferon alfa treatment could prolong the survival time of patients with HGG, especially the MGMT promoter unmethylation variant, and the toxic effects remained tolerable., Trial Registration: ClinicalTrials.gov Identifier: NCT01765088.
- Published
- 2023
- Full Text
- View/download PDF
46. Temozolomide-induced vitiligo-like reaction: An extremely rare phenomenon.
- Author
-
Madan R, Raja MK, Mahajan R, and Goyal S
- Subjects
- Humans, Middle Aged, Temozolomide pharmacology, Dacarbazine adverse effects, Antineoplastic Agents, Alkylating adverse effects, Vitiligo chemically induced, Vitiligo diagnosis, Vitiligo drug therapy, Brain Neoplasms drug therapy, Brain Neoplasms pathology, Glioblastoma drug therapy, Glioblastoma pathology
- Abstract
Abstract: Temozolomide (TMZ) is an alkylating chemotherapeutic drug, often used in high-grade glioma and metastatic melanoma. Common side effects of this include myelosuppression and gastrointestinal side effects. Vitiligo-like reaction is extremely rare after TMZ. Here, we report a case of a 55-year-old patient of glioblastoma who developed vitiligo-like reaction after starting adjuvant TMZ., (Copyright © 2023 Copyright: © 2023 Journal of Cancer Research and Therapeutics.)
- Published
- 2023
- Full Text
- View/download PDF
47. Evaluation of the Efficacy of Lactobacillus acidophilus in the Treatment of Cyclophosphamide-induced Hemorrhagic Cystitis in Wistar Rats.
- Author
-
Minhos LF, da Silva de Lima Gehlen AC, Júnior FFB, Dos Santos AC, Souza RIC, and de Barros ME
- Subjects
- Female, Rats, Animals, Rats, Wistar, Lactobacillus acidophilus, Antineoplastic Agents, Alkylating adverse effects, Cyclophosphamide adverse effects, Hemorrhage chemically induced, Hemorrhage drug therapy, Hemorrhage prevention & control, Inflammation drug therapy, Mesna adverse effects, Cystitis chemically induced, Cystitis drug therapy, Cystitis pathology
- Abstract
Background: Hemorrhagic cystitis is an inflammatory complication that can be caused by the administration of cyclophosphamide, which is widely used as an antineoplastic agent. In the search for new therapeutic alternatives, probiotics can suppress the inflammatory process and, therefore, can be used to prevent this disease., Objective: Thus, this study aimed to evaluate the effects of using Lactobacillus acidophilus NCFM in the treatment of cyclophosphamide-induced hemorrhagic cystitis in Wistar rats., Methods: Lactobacillus acidophilus NCFM (2x10
8 CFU) was used in the treatment of cyclophosphamide- induced hemorrhagic cystitis (200 mg/kg, intraperitoneal) in 77 female Wistar rats. Rats were distributed into experimental groups (n = 9): control group (GC), zero control group (GCZ), inflammation group (GI), 24-hour acute treatment groups: 24-hour lactobacilli treatment group (GL24H) and mesna group (GM), and 30-day chronic treatment groups: lactobacilli treatment group (GTL) and mesna+lactobacilli group (GM+L). After treatment, animals were euthanized and biological materials were collected for blood count, biochemical analyses, examination of abnormal sediment elements (EAS), and histopathological analysis., Results: GI results showed development of edema, macroscopic alterations, and signs of bleeding in the bladder; in addition, lesions in the urothelium and hemorrhage were also found. GL24H and GM presented intact urothelium, without inflammatory reaction and hematological or biochemical urine alterations., Conclusion: Therefore, this study demonstrated that L. acidophilus presented uroprotective effect against the action of cyclophosphamide in both the short and long term., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)- Published
- 2023
- Full Text
- View/download PDF
48. Feasibility of preirradiation temozolomide in cases of high-grade gliomas: Our experience and review of literature.
- Author
-
Arora S, Rathi AK, Singh K, and Ansari FA
- Subjects
- Humans, Temozolomide therapeutic use, Dacarbazine, Antineoplastic Agents, Alkylating adverse effects, Prospective Studies, Feasibility Studies, Brain Neoplasms drug therapy, Brain Neoplasms radiotherapy, Glioma drug therapy, Glioma radiotherapy, Glioblastoma, Astrocytoma chemically induced, Astrocytoma drug therapy
- Abstract
Aim: The purpose of this study was to evaluate the efficacy and safety of preradiation temozolomide (TMZ) in high-grade gliomas., Study and Design: It is a single-center, single arm, prospective study. The study included postoperative, histopatholgically proven cases of high-grade gliomas., Materials and Methods: Nine patients of anaplastic astrocytoma (AA) and twenty patients of glioblastoma multiforme (GBM) were enrolled in the study. All patients had undergone partial or complete resection. Three weeks after surgery, patients were started on chemotherapy, consisting of two cycles of TMZ, 150 mg/m
2 /day for 5 days, repeated at an interval of 4 weeks. Patients were subsequently treated with concomitant chemoradiotherapy. A dose of 60 Gy was given over thirty fractions along with TMZ, 75 mg/m2 /day. Four cycles of TMZ were given after completion of radiotherapy, in a dose and manner similar to preradiotherapy., Statistical Analysis and Result: Treatment-related toxicity was assessed using common terminology for toxicity criteria (CTCAE v4). Progression-free survival and overall survival (OS) analysis was done. Nearly 79% of patients completed the two cycles of preradiation chemotherapy. Chemotherapy was tolerated well. Median time to progression was 11 months and 8.2 months in AA and GBM patients, respectively. Median OS was 17.4 months in AA patients and 11.4 months in GBM patients., Conclusions: Most patients of postoperative high-grade gliomas tolerated two cycles of TMZ. A good safety profile of TMZ allows it to be used in frontline settings, especially in high volume centers where a delay in starting radiotherapy frequently occurs. The use of TMZ before radiotherapy is a safe and feasible approach, and further studies are required to validate this approach.- Published
- 2023
- Full Text
- View/download PDF
49. Patterns, predictors and prognostic relevance of high-grade hematotoxicity after temozolomide or temozolomide-lomustine in the CeTeG/NOA-09 trial.
- Author
-
Weller J, Schäfer N, Schaub C, Tzaridis T, Zeyen T, Schneider M, Potthoff AL, Giordano FA, Steinbach JP, Zeiner PS, Kowalski T, Sabel M, Hau P, Krex D, Grauer O, Goldbrunner R, Schnell O, Tabatabai G, Ringel F, Schmidt-Graf F, Brehmer S, Tonn JC, Bullinger L, Vajkoczy P, Glas M, Vatter H, Herrlinger U, and Seidel C
- Subjects
- Humans, Female, Temozolomide therapeutic use, Lomustine therapeutic use, Prognosis, Dacarbazine adverse effects, Antineoplastic Agents, Alkylating adverse effects, Brain Neoplasms therapy, Glioblastoma therapy
- Abstract
Purpose: In the randomized phase III trial CeTeG/NOA-09, temozolomide (TMZ)/lomustine (CCNU) combination therapy was superior to TMZ in newly diagnosed MGMT methylated glioblastoma, albeit reporting more frequent hematotoxicity. Here, we analyze high grade hematotoxicity and its prognostic relevance in the trial population., Methods: Descriptive and comparative analysis of hematotoxicity adverse events ≥ grade 3 (HAE) according to the Common Terminology of Clinical Adverse Events, version 4.0 was performed. The association of HAE with survival was assessed in a landmark analysis. Logistic regression analysis was performed to predict HAE during the concomitant phase of chemotherapy., Results: HAE occurred in 36.4% and 28.6% of patients under CCNU/TMZ and TMZ treatment, respectively. The median onset of the first HAE was during concomitant chemotherapy (i.e. first CCNU/TMZ course or daily TMZ therapy), and 42.9% of patients with HAE receiving further courses experienced repeat HAE. Median HAE duration was similar between treatment arms (CCNU/TMZ 11.5; TMZ 13 days). Chemotherapy was more often discontinued due to HAE in CCNU/TMZ than in TMZ (19.7 vs. 6.3%, p = 0.036). The occurrence of HAE was not associated with survival differences (p = 0.76). Regression analysis confirmed older age (OR 1.08) and female sex (OR 2.47), but not treatment arm, as predictors of HAE., Conclusion: Older age and female sex are associated with higher incidence of HAE. Although occurrence of HAE was not associated with shorter survival, reliable prediction of patients at risk might be beneficial to allow optimal management of therapy and allocation of supportive measures., Trial Registration: NCT01149109., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
50. Efficacy and safety of trabectedin for the treatment of advanced uterine or ovarian carcinosarcoma: Results of a phase II multicenter clinical trial (MITO-26).
- Author
-
Lorusso D, Pignata S, Tamberi S, Mangili G, Bologna A, Nicoloso MS, Giolitto S, Salutari V, Mantero M, Pisano C, Bergamini A, Musacchio L, Ronzulli D, Raspagliesi F, and Scambia G
- Subjects
- Adult, Female, Humans, Trabectedin adverse effects, Dioxoles adverse effects, Progression-Free Survival, Antineoplastic Agents, Alkylating adverse effects, Disease-Free Survival, Tetrahydroisoquinolines adverse effects, Ovarian Neoplasms drug therapy, Ovarian Neoplasms chemically induced, Uterine Neoplasms drug therapy, Uterine Neoplasms chemically induced
- Abstract
Objective: This open-label phase II clinical trial evaluated the antitumor activity and safety of trabectedin in patients with advanced ovarian (OC) or uterine carcinosarcomas (UC)., Methods: Eligible patients were adults (≥18 years) with histologically proven recurrent OC/UC not amenable to surgery or radiotherapy who received up to two prior chemotherapy lines. Trabectedin 1.3 mg/m
2 was administered as a 3-h infusion every three weeks. The primary endpoint was objective response rate (ORR) as per RECIST v.1.1. If at least 8 of 43 patients (18.6%) achieve an objective response, trabectedin would be declared worthy for further investigations., Results: Forty-five patients with either OC (n = 32) or UC (n = 13) from seven MITO centers across Italy were enrolled. The ORR was 11.9% (90% CI: 6-23) and included two patients with a complete response and three with a partial response. Eight patients (19.0%) had disease stabilization for a disease control rate of 31.0% (90% CI: 20-44). Median progression-free survival was 2.01 months (95% CI: 1.78-2.30) and median overall survival was 4.64 months (95% CI: 3.19-8.29). Neutrophil count decreases (n = 8, 18.2%) and transaminase increases (n = 6, 13.6%) were the most common grade 3-5 adverse events related with trabectedin. Two patients died due to trabectedin-related grade 5 hematological toxicity., Conclusion: Although trabectedin did not meet the prespecified activity criteria, it confers modest but clinically meaningful benefit to patients with advanced OC/UC as being as effective as any other available treatment for this indication. The toxicity profile appears in line with that previously reported for the drug., Competing Interests: Declaration of Competing Interest DL reports personal fees, travel support and institutional grants and founding from PharmaMar. SP reports personal fees from MSD, AstraZeneca, Roche, PharmaMar, Clovis and GSK; research funding from AstraZeneca, MSD, Roche and Pfizer. VS reports personal fees from MSD, GSK, Roche, PharmaMar, Clovis, Eisai and AstraZeneca; travel support from Roche, PharmaMar and AstraZeneca; funding from MSD, GSK, Roche and AstraZeneca. AB (Alice Bergamini) reports personal fees from GSK, PharmaMar, Clovis, Eisai and AstraZeneca/MSD; funding from GSK and AstraZeneca/MSD. All remaining authors have declared no conflicts of interest., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.