7,682 results on '"Antimetabolites, Antineoplastic"'
Search Results
2. Pevonedistat plus azacitidine vs azacitidine alone in higher-risk MDS/chronic myelomonocytic leukemia or low-blast-percentage AML
- Author
-
Lionel Adès, Larisa Girshova, Vadim A. Doronin, María Díez-Campelo, David Valcárcel, Suman Kambhampati, Nora-Athina Viniou, Dariusz Woszczyk, Raquel De Paz Arias, Argiris Symeonidis, Achilles Anagnostopoulos, Eduardo Ciliao Munhoz, Uwe Platzbecker, Valeria Santini, Robert J. Fram, Ying Yuan, Sharon Friedlander, Douglas V. Faller, Mikkael A. Sekeres, Institut Català de la Salut, [Adès L] INSERM U944, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint Louis and University of Paris, Paris, France. [Girshova L] Federal Almazov North-West Medical Research Centre, Saint-Petersburg, Russia. [Doronin VA] City Clinical Hospital 40, Moscow, Russia. [Díez-Campelo M] Institute for Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, Salamanca, Spain. [Valcárcel D] Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Barcelona, Spain. [Kambhampati S] Sarah Cannon at Research Medical Center, Kansas City, MO, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
acciones y usos químicos::acciones farmacológicas::mecanismos moleculares de acción farmacológica::antimetabolitos::antimetabolitos antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,Antimetabolites, Antineoplastic ,terapéutica::terapéutica::farmacoterapia::protocolos antineoplásicos::terapéutica::farmacoterapia::protocolos de quimioterapia antineoplásica combinada [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Chemical Actions and Uses::Pharmacologic Actions::Molecular Mechanisms of Pharmacological Action::Antimetabolites::Antimetabolites, Antineoplastic [CHEMICALS AND DRUGS] ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Leukemia, Myelomonocytic, Chronic ,neoplasias::neoplasias por tipo histológico::leucemia::leucemia mieloide::leucemia mielomonocítica crónica [ENFERMEDADES] ,Cyclopentanes ,Hematology ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Otros calificadores::Otros calificadores::/efectos adversos [Otros calificadores] ,Quimioteràpia combinada ,Medicaments antineoplàstics - Efectes secundaris ,Neoplasms::Neoplasms by Histologic Type::Leukemia::Leukemia, Myeloid::Leukemia, Myelomonocytic, Chronic [DISEASES] ,Pyrimidines ,Therapeutics::Therapeutics::Drug Therapy::Antineoplastic Protocols::Therapeutics::Drug Therapy::Antineoplastic Combined Chemotherapy Protocols [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,Leucèmia mieloide aguda - Tractament ,Other subheadings::Other subheadings::/adverse effects [Other subheadings] ,Azacitidine ,Humans ,Drug Therapy, Combination - Abstract
Pevonedistat; Chronic myelomonocytic leukemia Pevonedistat; Leucemia mielomonocítica crónica Pevonedistat; Leucèmia mielomonocítica crònica PANTHER is a global, randomized phase 3 trial of pevonedistat+azacitidine (n = 227) vs azacitidine monotherapy (n = 227) in patients with newly diagnosed higher-risk myelodysplastic syndromes (MDS; n = 324), higher-risk chronic myelomonocytic leukemia (n = 27), or acute myeloid leukemia (AML) with 20% to 30% blasts (n = 103). The primary end point was event-free survival (EFS). In the intent-to-treat population, the median EFS was 17.7 months with pevonedistat+azacitidine vs 15.7 months with azacitidine (hazard ratio [HR], 0.968; 95% confidence interval [CI], 0.757-1.238; P = .557) and in the higher-risk MDS cohort, median EFS was 19.2 vs 15.6 months (HR, 0.887; 95% CI, 0.659-1.193; P = .431). Median overall survival (OS) in the higher-risk MDS cohort was 21.6 vs 17.5 months (HR, 0.785; P = .092), and in patients with AML with 20% to 30% blasts was 14.5 vs 14.7 months (HR, 1.107; P = .664). In a post hoc analysis, median OS in the higher-risk MDS cohort for patients receiving >3 cycles was 23.8 vs 20.6 months (P = .021) and for >6 cycles was 27.1 vs 22.5 months (P = .008). No new safety signals were identified, and the azacitidine dose intensity was maintained. Common hematologic grade ≥3 treatment emergent adverse events were anemia (33% vs 34%), neutropenia (31% vs 33%), and thrombocytopenia (30% vs 30%). These results underscore the importance of large, randomized controlled trials in these heterogeneous myeloid diseases and the value of continuing therapy for >3 cycles. The trial was registered on clinicaltrials.gov as #NCT03268954. This study was sponsored by Takeda Development Centers Inc (TDCA; Lexington, MA).
- Published
- 2022
3. Impact of Hypomethylating Agent Use on Hospital and Emergency Room Visits, and Predictors of Early Discontinuation in Patients With Higher-Risk Myelodysplastic Syndromes
- Author
-
Amer M. Zeidan, Namita Joshi, Hrishikesh Kale, Wei-Jhih Wang, Shelby Corman, Tehseen Salimi, and Robert S. Epstein
- Subjects
Antimetabolites, Antineoplastic ,Cancer Research ,Hematology ,DNA Methylation ,Decitabine ,Medicare ,Hospitals ,United States ,Treatment Outcome ,Oncology ,Myelodysplastic Syndromes ,Azacitidine ,Humans ,Emergency Service, Hospital ,Aged - Abstract
Previous analyses using the SEER-Medicare database have reported substantial underutilization of hypomethylating agents (HMAs) among patients with higher-risk myelodysplastic syndromes (MDS), and an association between poor HMA persistence and high economic burden. We aimed to compare rates of hospitalizations and emergency room (ER) visits among patients with higher-risk MDS according to use or non-use of HMA therapy, and to explore factors associated with early discontinuation of HMA therapy.We used the 2010-2016 SEER-Medicare database to identify patients aged ≥66 years with a new diagnosis of refractory anemia with excess blasts (RAEB; a surrogate for higher-risk MDS) between 2011 and 2015. New hospitalizations and ER visits during the 12 months following MDS diagnosis were determined. Treatment discontinuation was defined as stopping HMA therapy before 4 cycles.Overall, 664 (55.8%) patients were HMA users and 526 (44.2%) non-users. Non-users had more hospitalizations (mean 0.47 vs. 0.30, P.001) and ER visits (mean 0.69 vs. 0.41, P = .005) per month than HMA users. Among HMA users, 193 (29.1%) discontinued HMA therapy before 4 cycles, and 91 (47.2%) of these after 1 cycle. Older age and poor performance status were associated with higher risk of HMA discontinuation.An increased rate of hospitalizations and ER visits occurred in HMA non-users vs. HMA users. Approximately one-third of patients discontinued HMA therapy early. Predictors of discontinuation included older age and poor performance status. Novel approaches are needed to improve utilization and persistence with HMA therapy and associated outcomes, particularly among these higher-risk groups.
- Published
- 2022
4. Survey of US Medical Oncologists' Practices and Beliefs Regarding
- Author
-
Kyoin, Koo, Amy L, Pasternak, N Lynn, Henry, Vaibhav, Sahai, and Daniel L, Hertz
- Subjects
Oncologists ,Antimetabolites, Antineoplastic ,Dihydropyrimidine Dehydrogenase Deficiency ,Humans ,Breast Neoplasms ,Female ,Fluorouracil ,ORIGINAL CONTRIBUTIONS ,Capecitabine ,Dihydrouracil Dehydrogenase (NADP) - Abstract
PURPOSE: Patients who carry reduced-activity DPYD polymorphisms have increased fluoropyrimidine (FP) toxicity risk. Although pretreatment DPYD testing is recommended throughout most of Europe, it is not recommended in the United States, and adoption has been limited. The objective of this survey was to describe the current practice in the United States regarding pretreatment DPYD testing and understand the factors deterring oncologists from ordering testing. METHODS: Survey invitations were e-mailed to 325 medical oncologists practicing in the United States who are members of the SWOG Cancer Research Network Gastrointestinal Cancer, Breast Cancer, or Early Therapeutics Committees. Descriptive statistics were used to evaluate survey responses. RESULTS: Responses were collected from 59 (18.2%) US medical oncologists, of whom 98% strongly or somewhat agree that patients with dihydropyrimidine dehydrogenase (DPD) deficiency have increased toxicity risk and 96% would modify FP dosing for a patient with known DPD deficiency. However, only 32% strongly or somewhat agree that pretreatment DPYD testing is useful to inform FP treatment, 20% have ever ordered pretreatment testing, and 3% order testing for at least 10% of their FP-treated patients. The most important factors that deter oncologists from ordering testing were low prevalence of DPD deficiency (54%) and lack of clinical practice guideline recommendations (48%). CONCLUSION: Clinical adoption of pretreatment DPYD testing is extremely limited in the United States. Utilization may be substantially increased by inclusion in the oncology clinical practice guideline recommendations, coverage through health insurance, and potentially education of medical oncologists regarding available treatment modification guidelines.
- Published
- 2023
5. Prospective comparison of outcomes with azacitidine and decitabine in patients with AML ineligible for intensive chemotherapy
- Author
-
Amer M. Zeidan, Pierre Fenaux, Marco Gobbi, Jiří Mayer, Gail J. Roboz, Jürgen Krauter, Tadeusz Robak, Hagop M. Kantarjian, Jan Novák, Wieslaw W. Jedrzejczak, Xavier Thomas, Mario Ojeda-Uribe, Yasushi Miyazaki, Yoo Hong Min, Su-Peng Yeh, Joseph M. Brandwein, Liana Gercheva, Judit Demeter, Elizabeth A. Griffiths, Karen W. L. Yee, Jean-Pierre J. Issa, Jan Philipp Bewersdorf, Harold Keer, Yong Hao, Mohammad Azab, and Hartmut Döhner
- Subjects
Antimetabolites, Antineoplastic ,Leukemia, Myeloid, Acute ,Treatment Outcome ,Antineoplastic Combined Chemotherapy Protocols ,Immunology ,Azacitidine ,Humans ,Cell Biology ,Hematology ,Decitabine ,Biochemistry - Published
- 2022
6. Association between Weight Gain and Sex-Related Differences through 5-Fluorouracil Administration.
- Author
-
Imai M, Hiramoto K, Tanaka S, and Ooi K
- Subjects
- Animals, Female, Male, Antimetabolites, Antineoplastic, Eating drug effects, Mice, Estradiol blood, Norepinephrine metabolism, Mice, Inbred C57BL, Fluorouracil, Weight Gain drug effects, Sex Characteristics, Ghrelin
- Abstract
Research on sex differences has increased across various fields, including cancer and its treatment domains. Reports have indicated sex differences in cancer incidence, survival rates, and the efficacy of anticancer drugs. However, such reports are limited, and in-depth assessments of the underlying mechanisms are still in progress. Although various chemotherapeutic regimens are applicable for breast cancer treatment, reports have surfaced regarding weight gain in female patients undergoing fluorouracil, epirubicin, cyclophosphamide (FEC) or cyclophosphamide, methotrexate, fluorouracil (CMF) therapy. We hypothesized the potential of 5-fluorouracil (5-FU) in weight gain and sex-related differences. To address this, we conducted experiments in mice to confirm weight gain and sex differences following 5-FU administration, and elucidate the underlying mechanisms. Our findings revealed weight gain and increased food intake in female mice following 5-FU administration. Additionally, female mice receiving 5-FU exhibited increased norepinephrine and α1- and α2-adrenergic receptor expression, reduced estradiol levels, and increased ghrelin levels. These results indicate 5-FU administration-induced sex differences in weight gain and implicate increased food intake because of increased norepinephrine and α1- and α2-adrenergic receptor expression, reduced estradiol levels, and a subsequent increase in ghrelin levels, which contribute to weight gain in female patients undergoing CMF therapy.
- Published
- 2024
- Full Text
- View/download PDF
7. Updated DPYD HapB3 haplotype structure and implications for pharmacogenomic testing.
- Author
-
Turner AJ, Haidar CE, Yang W, Boone EC, Offer SM, Empey PE, Haddad A, Tahir S, Scharer G, Broeckel U, and Gaedigk A
- Subjects
- Child, Humans, Haplotypes, Antimetabolites, Antineoplastic, Pharmacogenomic Testing, Genotype, Dihydrouracil Dehydrogenase (NADP) metabolism, Population Health
- Abstract
The DPYD gene encodes dihydropyrimidine dehydrogenase, the rate-limiting enzyme for the metabolism of fluoropyrimidines 5-fluorouracil and capecitabine. Genetic variants in DPYD have been associated with altered enzyme activity, therefore accurate detection and interpretation is critical to predict metabolizer status for individualized fluoropyrimidine therapy. The most commonly observed deleterious variation is the causal variant linked to the previously described HapB3 haplotype, c.1129-5923C>G (rs75017182) in intron 10, which introduces a cryptic splice site. A benign synonymous variant in exon 11, c.1236G>A (rs56038477) is also linked to HapB3 and is commonly used for testing. Previously, these single-nucleotide polymorphisms (SNPs) have been reported to be in perfect linkage disequilibrium (LD); therefore, c.1236G>A is often utilized as a proxy for the function-altering intronic variant. Clinical genotyping of DPYD identified a patient who had c.1236G>A, but not c.1129-5923C>G, suggesting that these two SNPs may not be in perfect LD, as previously assumed. Additional individuals with c.1236G>A, but not c.1129-5923C>G, were identified in the Children's Mercy Data Warehouse and the All of Us Research Program version 7 cohort substantiating incomplete SNP linkage. Consequently, testing only c.1236G>A can generate false-positive results in some cases and lead to suboptimal dosing that may negatively impact patient therapy and prospect of survival. Our data show that DPYD genotyping should include the functional variant c.1129-5923C>G, and not the c.1236G>A proxy, to accurately predict DPD activity., (© 2023 The Authors. 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
8. Oral Azacitidine (CC-486) for the Treatment of Myeloid Malignancies
- Author
-
C.L. Beach, Hartmut Döhner, Valeria Santini, Andrew H. Wei, Ignazia La Torre, Guillermo Garcia-Manero, and Barry Skikne
- Subjects
Adult ,Oncology ,Antimetabolites, Antineoplastic ,Cancer Research ,medicine.medical_specialty ,Combination therapy ,law.invention ,Maintenance therapy ,Randomized controlled trial ,law ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Immunologic Factors ,media_common.cataloged_instance ,European union ,neoplasms ,Randomized Controlled Trials as Topic ,media_common ,Myeloproliferative Disorders ,business.industry ,Myelodysplastic syndromes ,Hematopoietic Stem Cell Transplantation ,Induction chemotherapy ,Hematology ,medicine.disease ,Clinical trial ,Leukemia, Myeloid, Acute ,stomatognathic diseases ,Clinical Trials, Phase III as Topic ,Hypomethylating agent ,Myelodysplastic Syndromes ,Azacitidine ,business - Abstract
Epigenetic dysregulation leads to aberrant DNA hypermethylation and is common in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). A large number of clinical trials in AML, MDS, and other hematologic malignancies have assessed hypomethylating agents (HMAs), used alone or in combination with other drugs, in the frontline, maintenance, relapsed/refractory, and peritransplant settings. Effective maintenance therapy has long been a goal for patients with AML in remission. Previous large, randomized clinical trials of maintenance with HMAs or other agents had not shown meaningful improvement in overall survival. Oral azacitidine (Oral-AZA [CC-486]) is approved in the United States, Canada, and European Union for treatment of adult patients with AML in first complete remission (CR) or CR with incomplete blood count recovery (CRi) following intensive induction chemotherapy who are ineligible for hematopoietic cell transplant. Regulatory approvals of Oral-AZA were based on outcomes from the randomized, phase III QUAZAR AML-001 trial, which showed a median overall survival advantage of 9.9 months with Oral-AZA versus placebo. Oral-AZA allows convenient extended AZA dosing for 14 days per 28-day treatment cycle, which is not feasible with injectable AZA. Focusing on AML and MDS, this report reviews the rationale for the use of orally bioavailable AZA and its potential use in all-oral combination therapy regimens; the unique pharmacokinetic and pharmacodynamic profile of Oral-AZA compared with injectable AZA; the clinical safety and efficacy of Oral-AZA maintenance therapy in patients with AML in first remission and for treatment of patients with active MDS; and ongoing Oral-AZA clinical trials.
- Published
- 2022
9. Chidamide and Decitabine in Combination with a HAG Priming Regimen for Acute Myeloid Leukemia with TP53 Mutation
- Author
-
Bei, Zhang, Zhixin, Pei, Hongxia, Wang, Huimin, Wu, Junjie, Wang, Junjun, Bai, and Qinglin, Song
- Subjects
Adult ,Male ,Antimetabolites, Antineoplastic ,HAG ,Remission Induction ,Cytarabine ,TP53 mutation ,Aminopyridines ,Middle Aged ,acute myeloid leukemia ,Leukemia, Myeloid, Acute ,Treatment Outcome ,chidamide ,Benzamides ,Granulocyte Colony-Stimulating Factor ,Mutation ,Humans ,Female ,Tumor Suppressor Protein p53 ,Homoharringtonine ,decitabine ,Aged ,Retrospective Studies - Abstract
We analyzed the treatment effects of chidamide and decitabine in combination with a HAG (homoharringtonine, cytarabine, G-CSF) priming regimen (CDHAG) in acute myeloid leukemia (AML) patients with TP53 mutation. Seven TP53 mutated AML patients were treated with CDHAG. The treatment effects were assessed using hemogram detection and bone marrow aspirate. The possible side effects were evaluated based on both hematological and non-hematological toxicity. Four of the seven patients were classified as having achieved complete remission after CDHAG treatment; one patient was considered to have achieved partial remission, and the remaining two patients were considered in non-remission. The overall response rate (ORR) to CDHAG was 71.4%. Regarding the side effects, the hematological toxicity level of the seven patients ranged from level III to level IV, and infections that occurred at lung, blood, and skin were recorded. Nausea, vomiting, liver injury, and kidney injury were also detected. However, all side effects were attenuated by proper management. The CDHAG regimen clearly improved the ORR (71.4%) of TP53-mutated AML patients, with no severe side effects.
- Published
- 2022
10. Effect of the Proton Pump Inhibitor Esomeprazole on the Systemic Exposure of Capecitabine
- Author
-
Peter de Bruijn, Sander Bins, Ferry A.L.M. Eskens, Femke M. de Man, Ron H.J. Mathijssen, Niels Heersche, Esther Oomen-de Hoop, Leni van Doorn, Ivo Bijl, Ate van der Gaast, and Medical Oncology
- Subjects
Male ,Antimetabolites, Antineoplastic ,medicine.medical_specialty ,medicine.drug_class ,Biological Availability ,Proton-pump inhibitor ,Carbonated Beverages ,Gastroenterology ,Esomeprazole ,Capecitabine ,Pharmacokinetics ,SDG 3 - Good Health and Well-being ,Neoplasms ,Internal medicine ,medicine ,Clinical endpoint ,Humans ,Drug Interactions ,Pharmacology (medical) ,Prospective Studies ,Netherlands ,Pharmacology ,Cross-Over Studies ,business.industry ,Proton Pump Inhibitors ,Middle Aged ,Drug interaction ,Crossover study ,Treatment Outcome ,Concomitant ,Female ,Drug Monitoring ,business ,medicine.drug - Abstract
Retrospective data suggest that gastric acid reduction by proton pump inhibitors (PPIs) impairs the dissolution and subsequent absorption of capecitabine, and thus potentially reduces the capecitabine exposure. Therefore, we examined prospectively the effect of esomeprazole on the pharmacokinetics of capecitabine. In this randomized crossover study, patients with cancer were assigned to 2 sequence groups, each consisting of 3 phases: capecitabine with esomeprazole administration 3 hours before (phase A), capecitabine alone (phase B), and capecitabine concomitant with cola and esomeprazole co-administration 3 hours before (phase C). The primary end point was the relative difference (RD) in exposure to capecitabine assessed by the area under the plasma concentration-time curve from zero to infinity (AUC0-inf) and analyzed by a linear mixed effect model. Twenty-two evaluable patients were included in the analysis. After esomeprazole, there was a 18.9% increase in AUC0-inf of capecitabine (95% confidence interval (CI) −10.0% to 57.0%, P = 0.36). In addition, capecitabine half-life was significantly longer after esomeprazole (median 0.63 hours vs. 0.46 hours, P = 0.005). Concomitant cola did not completely reverse the effects observed after esomeprazole (RD 3.3% (95% CI −16.3 to 27.4%, P = 1.00). Capecitabine exposure is not negatively influenced by esomeprazole cotreatment. Therefore, altered capecitabine pharmacokinetics do not explain the assumed worse clinical outcome of PPI-cotreated patients with cancer.
- Published
- 2022
11. Oral-recombinant Methioninase Converts an Osteosarcoma from Methotrexate-resistant to -sensitive in a Patient-derived Orthotopic-xenograft (PDOX) Mouse Model
- Author
-
YUSUKE AOKI, YASUNORI TOME, QINGHONG HAN, JUN YAMAMOTO, KAZUYUKI HAMADA, NORIYUKI MASAKI, YUTARO KUBOTA, MICHAEL BOUVET, KOTARO NISHIDA, and ROBERT M. HOFFMAN
- Subjects
Antimetabolites, Antineoplastic ,Osteosarcoma ,Cancer Research ,Administration, Oral ,Mice, Nude ,Bone Neoplasms ,General Medicine ,Xenograft Model Antitumor Assays ,Tumor Burden ,Carbon-Sulfur Lyases ,Disease Models, Animal ,Mice ,Methotrexate ,Treatment Outcome ,Oncology ,Drug Resistance, Neoplasm ,Antineoplastic Combined Chemotherapy Protocols ,Animals ,Humans - Abstract
Osteosarcoma is the most common bone sarcoma. Although surgery and chemotherapy are initially effective, the 5-year survival is approximately 60% to 80%, and has not improved over three decades. We have previously shown that methionine restriction (MR) induced by oral recombinant methioninase (o-rMETase), is effective against osteosarcoma in patient-derived orthotopic xenograft (PDOX) nude-mouse models. In the present report, the efficacy of the combination of oral o-rMETase and methotrexate (MTX) was examined in an osteosarcoma PDOX mouse model.An osteosarcoma-PDOX model was previously established by implanting tumor fragments into the proximal tibia of nude mice. The osteosarcoma PDOX models were randomized into four groups: control; o-rMETase alone; MTX alone; combination of o-rMETase and MTX. The mice were sacrificed after 4 weeks of treatment.The combination of o-rMETase and MTX showed significantly higher efficacy compared to the control group (p=0.04). The combination also showed significantly higher efficacy compared to MTX alone (p=0.04). No significant efficacy of o-rMETase alone or MTX alone compared to control was shown (p=0.21, 1.00, respectively). Only the combination of o-rMETase and MTX reduced the cancer-cell density in the osteosarcoma tumor.rMETase converted an osteosarcoma PDOX from MTX-resistant to MTX-sensitive and thereby shows future clinical potential.
- Published
- 2022
12. Methionine Restriction: Ready for Prime Time in the Cancer Clinic?
- Author
-
Jun, Yamamoto, Qinghong, Han, Mark, Simon, Daniel, Thomas, and Robert M, Hoffman
- Subjects
Antimetabolites, Antineoplastic ,Carbon-Sulfur Lyases ,Cancer Research ,Methionine ,Oncology ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Diet, Protein-Restricted ,Animals ,Humans ,General Medicine - Abstract
Attempts to selectively starve cancers in the clinic have been made at least since the time of Warburg beginning 100 years ago. Calorie-restriction or low-carbohydrate diets have had limited success with cancer patients. Methionine restriction is another strategy to selectively starve cancer cells, since cancers are addicted to methionine, unlike normal cells. Methionine addiction of cancer is termed the Hoffman effect. Numerous preclinical studies over the past half century have shown methionine restriction to be highly effective against all major cancer types and synergistic with chemotherapy. Low-methionine medical diets can be effective in lowering methionine and have shown some clinical promise, but they are not palatable and thereby not sustainable. However, selectively choosing among plant-based foods allows a variety of low-methionine diets that are sustainable. Our laboratory has developed a methioninase that can be administered orally as a supplement and has resulted in anecdotal positive results in patients with advanced cancer, including hormone-independent prostate cancer, and other recalcitrant cancers. The question is whether methionine restriction through a low-methionine diet, or even greater methionine restriction with methioninase in combination with a low-methionine diet, is ready for prime time in the clinic, especially in combination with other synergistic therapy. The question will hopefully be answered in the near future, especially for advanced cancer patients who have failed all standard therapy.
- Published
- 2022
13. Azacitidine-induced reconstitution of the bone marrow T cell repertoire is associated with superior survival in AML patients
- Author
-
Grimm, Juliane, Simnica, Donjete, Jäkel, Nadja, Paschold, Lisa, Willscher, Edith, Schulze, Susann, Dierks, Christine, Al-Ali, Haifa Kathrin, and Binder, Mascha
- Subjects
Aged, 80 and over ,Male ,Antimetabolites, Antineoplastic ,T-Lymphocytes ,Immunosurveillance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Middle Aged ,Survival Analysis ,Article ,Acute myeloid leukaemia ,Leukemia, Myeloid, Acute ,Bone Marrow ,Azacitidine ,Humans ,T-cell receptor ,Female ,RC254-282 ,Aged - Abstract
Hypomethylating agents (HMA) like azacitidine are licensed for the treatment of acute myeloid leukemia (AML) patients ineligible for allogeneic hematopoietic stem cell transplantation. Biomarker-driven identification of HMA-responsive patients may facilitate the choice of treatment, especially in the challenging subgroup above 60 years of age. Since HMA possesses immunomodulatory functions that constitute part of their anti-tumor effect, we set out to analyze the bone marrow (BM) immune environment by next-generation sequencing of T cell receptor beta (TRB) repertoires in 51 AML patients treated within the RAS-AZIC trial. Patients with elevated pretreatment T cell diversity (11 out of 41 patients) and those with a boost of TRB richness on day 15 after azacitidine treatment (12 out of 46 patients) had longer event-free and overall survival. Both pretreatment and dynamic BM T cell metrics proved to be better predictors of outcome than other established risk factors. The favorable broadening of the BM T cell space appeared to be driven by antigen since these patients showed significant skewing of TRBV gene usage. Our data suggest that one course of AZA can cause reconstitution to a more physiological T cell BM niche and that the T cell space plays an underestimated prognostic role in AML. Trial registration: DRKS identifier: DRKS00004519
- Published
- 2022
14. Prospective evaluation of high‐dose methotrexate pharmacokinetics in adult patients with lymphoma using novel determinants of kidney function
- Author
-
Andrew D. Rule, Kianoush Kashani, Carrie A. Thompson, Joel M. Reid, Thomas E. Witzig, Nelson Leung, Renee M. McGovern, Kristin C. Mara, Erin F. Barreto, Thomas R. Larson, and Jason N. Barreto
- Subjects
Male ,musculoskeletal diseases ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,Lymphoma ,Population ,Urology ,Renal function ,RM1-950 ,Kidney Function Tests ,General Biochemistry, Genetics and Molecular Biology ,Article ,chemistry.chemical_compound ,Pharmacokinetics ,Interquartile range ,medicine ,Humans ,Prospective Studies ,General Pharmacology, Toxicology and Pharmaceutics ,education ,Aged ,education.field_of_study ,Creatinine ,business.industry ,General Neuroscience ,Research ,Area under the curve ,General Medicine ,Articles ,Middle Aged ,Methotrexate ,chemistry ,Female ,Liver function ,Therapeutics. Pharmacology ,Public aspects of medicine ,RA1-1270 ,business ,medicine.drug ,Glomerular Filtration Rate - Abstract
High‐dose methotrexate (HDMTX) pharmacokinetics (PKs), including the best estimated glomerular filtration rate (eGFR) equation that reflects methotrexate (MTX) clearance, requires investigation. This prospective, observational, single‐center study evaluated adult patients with lymphoma treated with HDMTX. Samples were collected at predefined time points up to 96 h postinfusion. MTX and 7‐hydroxy‐MTX PKs were estimated by standard noncompartmental analysis. Linear regression determined which serum creatinine‐ or cystatin C‐based eGFR equation best predicted MTX clearance. The 80 included patients had a median (interquartile range [IQR]) age of 68.6 years (IQR 59.2–75.6), 54 (67.5%) were men, and 74 (92.5%) were White. The median (IQR) dose of MTX was 7.6 (IQR 4.8–11.3) grams. Median clearance was similar across three dosing levels at 4.5–5.6 L/h and was consistent with linear PKs. Liver function, weight, age, sex, concomitant chemotherapy, and number of previous MTX doses did not impact clearance. MTX area under the curve (AUC) values varied over a fourfold range and appeared to increase in proportion to the dose. The eGFRcys (ml/min) equation most closely correlated with MTX clearance in both the entire cohort and after excluding outlier MTX clearance values (r = 0.31 and 0.51, respectively). HDMTX as a 4‐h infusion displays high interpatient pharmacokinetic variability. Population PK modeling to optimize MTX AUC attainment requires further evaluation. The cystatin C‐based eGFR equation most closely estimated MTX clearance and should be investigated for dosing and monitoring in adults requiring MTX as part of lymphoma management.
- Published
- 2022
15. Microbial metabolite restricts 5-fluorouracil-resistant colonic tumor progression by sensitizing drug transporters via regulation of FOXO3-FOXM1 axis
- Author
-
Sweta Ghosh, Rajbir Singh, Zachary Matthew Vanwinkle, Haixun Guo, Praveen Kumar Vemula, Ajay Goel, Bodduluri Haribabu, and Venkatakrishna Rao Jala
- Subjects
Antimetabolites, Antineoplastic ,Forkhead Box Protein M1 ,Forkhead Box Protein O3 ,Azoxymethane ,Medicine (miscellaneous) ,Gastrointestinal Microbiome ,Neoplasm Proteins ,Mice ,Coumarins ,Drug Resistance, Neoplasm ,Cell Line, Tumor ,Colonic Neoplasms ,ATP Binding Cassette Transporter, Subfamily G, Member 2 ,Animals ,Humans ,Fluorouracil ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) - Abstract
The survival rate of colorectal cancer patients is adversely affected by the selection of tumors resistant to conventional anti-cancer drugs such as 5-fluorouracil (5FU). Although there is mounting evidence that commensal gut microbiota is essential for effective colon cancer treatment, the detailed molecular mechanisms and the role of gut microbial metabolites remain elusive. The goal of this study is to decipher the impact and mechanisms of gut microbial metabolite, urolithin A (UroA) and its structural analogue, UAS03 on reversal of 5FU-resistant (5FUR) colon cancers.
- Published
- 2022
16. Management of a complete hydatidiform mole with a coexisting live fetus followed by successful treatment of maternal metastatic gestational trophoblastic disease: learning points
- Author
-
Swee Lin Yip, Khurshid Merchant, and Zhun Wei Mok
- Subjects
Adult ,Male ,medicine.medical_specialty ,Down syndrome ,Antimetabolites, Antineoplastic ,Lung Neoplasms ,medicine.medical_treatment ,Case Report ,03 medical and health sciences ,0302 clinical medicine ,Obstetrics and gynaecology ,Breech presentation ,Pregnancy ,Placenta ,medicine ,Humans ,Gestational Trophoblastic Disease ,reproductive and urinary physiology ,Chemotherapy ,030219 obstetrics & reproductive medicine ,Obstetrics ,Gestational trophoblastic disease ,business.industry ,Infant, Newborn ,General Medicine ,Hydatidiform Mole ,medicine.disease ,medicine.anatomical_structure ,Methotrexate ,030220 oncology & carcinogenesis ,Uterine Neoplasms ,Female ,business ,medicine.drug - Abstract
A 34-year-old patient had her first trimester Down syndrome scan followed by serial ultrasound scans which showed a single intrauterine pregnancy with multiple cystic areas in the anterior placenta. She presented in preterm labour with a breech presentation at 32 weeks and underwent an emergency caesarean section. She delivered a male infant weighing 1750 g. The placental histopathology showed a complete hyatidiform mole. At 4 weeks postpartum, beta-human chorionic gonadotrophin (Bhcg) levels rose from 460 to 836 mIU/mL over 1 week. Metastatic workup revealed prominent pelvic nodes and pulmonary nodules in both lungs. This was discussed at the Multi-Disciplinary Tumour Board and single-agent intramuscular methotrexate was recommended. After chemotherapy, she achieved Bhcg normalisation after three cycles. This case highlights the importance of clinical vigilance even in low-risk patients. Unexpected findings on ultrasound should involve multidisciplinary input with radiology colleagues. A high index of suspicion for gestational trophoblastic disease and close follow-up is imperative.
- Published
- 2023
17. Integrin-Mediated Targeted Cancer Therapy Using c(RGDyK)-Based Conjugates of Gemcitabine
- Author
-
Theodora Chatzisideri, George Leonidis, Theodoros Karampelas, Eleni Skavatsou, Angeliki Velentza-Almpani, Francesca Bianchini, Constantin Tamvakopoulos, and Vasiliki Sarli
- Subjects
Antimetabolites, Antineoplastic ,Integrins ,Lung Neoplasms ,Deoxycytidine ,Peptides, Cyclic ,Gemcitabine ,Mice, Inbred C57BL ,Mice ,A549 Cells ,Integrin, c(RGDyK)-based conjugates, gemcitabine, Targeted Cancer Therapy ,Drug Discovery ,Animals ,Humans ,Molecular Medicine ,Cell Proliferation - Abstract
c(RGDyK)-based conjugates of gemcitabine (GEM) with the carbonate and carbamate linkages in the 6-OH group of GEM were synthesized for the targeted delivery of GEM to integrin α
- Published
- 2021
18. Oral hypomethylating agents: beyond convenience in MDS
- Author
-
Elizabeth A, Griffiths
- Subjects
Antimetabolites, Antineoplastic ,MDS: Beyond a One-Size-Fits-All Approach ,Myelodysplastic Syndromes ,Azacitidine ,Quality of Life ,Administration, Oral ,Humans ,Female ,Hematology ,Decitabine ,Uridine ,Aged - Abstract
Oral hypomethylating agents (HMAs) represent a substantial potential boon for patients with myelodysplastic syndrome (MDS) who have previously required between 5 and 7 visits per month to an infusion clinic to receive therapy. For patients who respond to treatment, ongoing monthly maintenance visits represent a considerable burden to quality of life, and for those who are early in therapy, these sequential visits may tax transportation and financial resources that would be optimally distributed over the treatment cycle to facilitate transfusion support. The availability of oral HMAs may support the optimal application of these agents by contributing to adherence and lessening the burden of therapy, potentially encouraging patients to stay on longer-term treatment. Distinct pharmacokinetic profiles for the recently approved oral HMAs (oral azacitidine and decitabine-cedazuridine) result in differential toxicity profiles and have prompted their clinical trial development in lower- and higher-risk MDS, respectively.
- Published
- 2021
19. High-dose AraC is essential for the treatment of ML-DS independent of postinduction MRD: results of the COG AAML1531 trial
- Author
-
R. Spencer Tong, Robert B. Gerbing, Todd A. Alonzo, Jeffrey W. Taub, Jim Wang, Lisa Eidenschink Brodersen, Reuven J. Schore, Maureen M. O'Brien, Amy Heerema-McKenney, Jason N. Berman, Shelton A Viola, Michael R. Loken, E. Anders Kolb, Betsy A. Hirsch, Alan S. Gamis, Karen M. Chisholm, Susana C. Raimondi, Nobuko Hijiya, Amy Beckman, Johann Hitzler, and Todd E. Druley
- Subjects
Male ,Antimetabolites, Antineoplastic ,medicine.medical_specialty ,Down syndrome ,Neoplasm, Residual ,medicine.medical_treatment ,Immunology ,Biochemistry ,Gastroenterology ,Cog ,Internal medicine ,medicine ,Humans ,Chemotherapy ,Dose-Response Relationship, Drug ,business.industry ,Cytarabine ,Infant ,Myeloid leukemia ,Cell Biology ,Hematology ,Prognosis ,Interim analysis ,medicine.disease ,Confidence interval ,Young age ,Treatment Outcome ,Leukemia, Myeloid ,Child, Preschool ,Female ,Down Syndrome ,business ,medicine.drug - Abstract
Myeloid leukemia in children with Down syndrome (ML-DS) is associated with young age and somatic GATA1 mutations. Because of high event-free survival (EFS) and hypersensitivity of the leukemic blasts to chemotherapy, the prior Children’s Oncology Group protocol ML-DS protocol (AAML0431) reduced overall treatment intensity but lacking risk stratification, retained the high-dose cytarabine course (HD-AraC), which was highly associated with infectious morbidity. Despite high EFS of ML-DS, survival for those who relapse is rare. AAML1531 introduced therapeutic risk stratification based on the previously identified prognostic factor, measurable residual disease (MRD) at the end of the first induction course. Standard risk (SR) patients were identified by negative MRD using flow cytometry (
- Published
- 2021
20. Secondary basophilic leukemia in Ph-negative myeloid neoplasms: A distinct subset with poor prognosis
- Author
-
Philipp B. Staber, Peter Bettelheim, Christian Sillaber, Gabriele Stefanzl, Dubravka Smiljkovic, Daniela Berger, Georg Greiner, Peter Valent, Karin Bauer, Wolfgang R. Sperr, Ana-Iris Schiefer, Susanne Gamperl, Ilse Schwarzinger, Renate Thalhammer, Gregor Hoermann, Paul Knöbl, and Christoph Kornauth
- Subjects
Male ,Antimetabolites, Antineoplastic ,Original article ,Cancer Research ,Myeloid ,Azacitidine ,chemical and pharmacologic phenomena ,Venetoclax ,chemistry.chemical_compound ,parasitic diseases ,medicine ,Humans ,IgE receptor ,Myeloproliferative neoplasm ,RC254-282 ,Aged ,Aged, 80 and over ,Acute leukemia ,Leukemia ,Myeloproliferative Disorders ,business.industry ,Myeloid leukemia ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Neoplasms, Second Primary ,hemic and immune systems ,Prognosis ,medicine.disease ,Basophilic leukemia ,Basophils ,Basophilia ,medicine.anatomical_structure ,chemistry ,Myelodysplastic Syndromes ,Cancer research ,Female ,business ,medicine.drug - Abstract
During progression of myeloid neoplasms, the basophil compartment may expand substantially and in some of these patients, a basophilic leukemia is diagnosed. In patients with Ph-chromosome+ chronic myeloid leukemia, acceleration of disease is typically accompanied by marked basophilia. In other myeloid neoplasms, secondary leukemic expansion of basophils is rarely seen. We report on 5 patients who suffered from a myelodysplastic syndrome, myeloproliferative neoplasm, or acute leukemia and developed a massive expansion of basophils during disease progression. In 4 of 5 patients, peripheral blood basophil counts reached 40%, and the diagnosis “secondary basophilic leukemia” was established. As assessed by flow cytometry, neoplastic basophils expressed CD9, CD18, CD25, CD33, CD63, PD-L1, CD123, and CLL-1. In addition, basophils were found to display BB1 (basogranulin), 2D7, tryptase and KIT. In 4 of 5 patients the disease progressed quickly and treatment with azacitidine was started. However, azacitidine did not induce major clinical responses, and all patients died from progressive disease within 3 Y. In in vitro experiments, the patients´ cells and the basophilic leukemia cell line KU812 showed variable responses to targeted drugs, including azacitidine, venetoclax, hydroxyurea, and cytarabine. A combination of venetoclax and azacitidine induced cooperative antineoplastic effects in these cells. Together, secondary basophilic leukemia has a poor prognosis and monotherapy with azacitidine is not sufficient to keep the disease under control for longer time-periods. Whether drug combination, such as venetoclax+azacitidine, can induce better outcomes in these patients remains to be determined in future clinical studies.
- Published
- 2021
21. Near Miss or Standard of Care? DPYD Screening for Cancer Patients Receiving Fluorouracil
- Author
-
Lauren E Winquist, Eric Winquist, Richard B. Kim, and Michael Sanatani
- Subjects
Oncology ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,Near Miss, Healthcare ,Case Report ,fluoropyrimidines ,030226 pharmacology & pharmacy ,Capecitabine ,03 medical and health sciences ,single nucleotide polymorphisms ,0302 clinical medicine ,Pharmacotherapy ,Internal medicine ,Neoplasms ,medicine ,Dihydropyrimidine dehydrogenase ,Humans ,cancer ,Dosing ,Adverse effect ,Dihydrouracil Dehydrogenase (NADP) ,Early Detection of Cancer ,RC254-282 ,Cancer ,business.industry ,Adverse effects ,dihydropyrimidine dehydrogenase ,Fluoropyrimidines ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Standard of Care ,Single nucleotide polymorphisms ,medicine.disease ,drug therapy ,Fluorouracil ,030220 oncology & carcinogenesis ,adverse effects ,DPYD ,Drug therapy ,business ,medicine.drug - Abstract
5-fluorouracil (5-FU) and its pro-drug capecitabine are widely used anticancer agents. Most 5-FU catabolism is dependent on dihydropyrimidine dehydrogenase (DPD) encoded by the DPYD gene, and DPYD variants that reduce DPD function increase 5-FU toxicity. Most DPD deficient patients are heterozygous and can be treated with reduced 5-FU dosing. We describe a patient with a genotype associated with near complete absence of DPD function, and severe and likely fatal toxicity with 5-FU treatment. The patient was treated effectively with alternative systemic therapy. Routine pretreatment DPYD genotyping is recommended by the European Medicines Agency, and guidelines for use of 5-FU in DPD deficient patients are available. However, outside the province of Quebec, routine pretreatment screening for DPD deficiency remains unavailable in Canada. It is likely our patient would have died from 5-FU toxicity under the current standard of care, but instead provides an example of the potential benefit of DPYD screening on patient outcomes.
- Published
- 2021
22. Quantitative detection of 6-thioguanine in body fluids based on a free-standing liquid membrane SERS substrate
- Author
-
Zhe Feng, Zhengjun Gong, Jing Liu, Sha-Na Zhou, Wen Liu, Meikun Fan, Xin Zhao, and Dongmei Wang
- Subjects
Detection limit ,Antimetabolites, Antineoplastic ,Silver ,Materials science ,Chromatography ,Calibration curve ,Metal Nanoparticles ,Substrate (chemistry) ,Spectrum Analysis, Raman ,Biochemistry ,Analytical Chemistry ,symbols.namesake ,Membrane ,Limit of Detection ,symbols ,Humans ,Gold ,Thioguanine ,Spectroscopy ,Raman spectroscopy ,Suspension (vehicle) ,6-Thioguanine - Abstract
The adverse reactions caused by 6-thioguanine (6-TG) in anti-cancer treatment are closely related to the dose, leading to the urgent need for clinical monitoring of its concentration. In this work, a highly reproducible free-standing liquid membrane (FLM) surface-enhanced Raman spectroscopy (SERS) substrate was developed to detect 6-TG in human urine and serum quantitatively. Briefly, a prepared sample was adjusted to pH 2 and mixed with concentrated core–shell bimetallic nanoparticle (AgcoreAushell NP) suspension. The Au/Ag ratio of the AgcoreAushell NPs was optimized. Then the mixture was formed into an FLM using a custom mold. The relative standard deviation (RSD) of the experimental results can be stabilized below 10% (n ≥ 10). The R2 of the calibration curve in the range of 10 ~ 100 μg kg−1 was 0.988. In addition, the limit of detection (LOD) (3σ/k) of 6-TG was 5 μg kg−1. The FLM SERS platform has been successfully applied to the rapid and reliable analysis of 6-TG spiked in body fluids.
- Published
- 2021
23. The Effects of Phyllanthus niruri Linn on Infiltrating Dendritic Cell and Ratio of Neutrophile/Lymphocytes in Chemotherapy of Sprague-Dawley Rats with Colorectal Cancer
- Author
-
Michael Tendean and Ignatius Riwanto
- Subjects
Male ,Antimetabolites, Antineoplastic ,Phyllanthus ,Neutrophils ,Colorectal cancer ,medicine.medical_treatment ,Lymphocyte ,Pharmacology ,Rats, Sprague-Dawley ,Lesion ,Capecitabine ,Leukocyte Count ,Immune system ,medicine ,Animals ,Lymphocytes ,Chemotherapy ,biology ,business.industry ,Dendritic Cells ,General Medicine ,Dendritic cell ,medicine.disease ,biology.organism_classification ,Rats ,Disease Models, Animal ,medicine.anatomical_structure ,medicine.symptom ,Colorectal Neoplasms ,business ,medicine.drug - Abstract
Background: Chemotherapy as part of colorectal cancer management can cause death to immunologically active tumor cell, but also it has immune suppressive effect. Phyllanthus niruri Linn is known to has immunomodulatory effect. This study was intended to prove P. niruri Linn effect on infiltrating dendritic cells and Neutrophil/lymphocyte ratios (NLRs) in Sprague–Dawley rats with colorectal cancer which were given capecitabine chemotherapy. Methods: The study was randomized post–test only control group design. The samples were 39 Sprague–Dawley male rats, with body weight around 170–220 grams, induced by 1,2-dimetylhydrazine (DMH) 30 mg/kgBW once per week subcutaneously. On 9th,11th and 13th week, there were four induced rats sacrificed each week to detect colorectal cancer (CRC) development. On the 13th week, all of the 4 sacrificed rats developed colon cancer, so the induction had to be stopped. The rest of 27 induced rats were randomly divided into three groups: control-group (K) were left untreated (9 rats), group P1 (9 rats) were given Capecitabine and group P2 (9 rats) were given Capecitabine with combination of P. niruri Linn extract 13.5 mg/kgBW orally. After 17th week, all rats were terminated and tumor lesion of colon were processed to be paraffin blocks and were stained with HE for evaluating the NLRs, and immunohistochemistry (S100) for evaluating infiltrating dendritic cells. Data was analyzed by using Oneway-Anova-test and post-Hoc LSD-test. Considered significant if p was
- Published
- 2021
24. Epigenetic activation of the elongator complex sensitizes gallbladder cancer to gemcitabine therapy
- Author
-
Xiaoqiang Hu, Xiaopeng Wang, Wei Chen, Tao Chen, Sunwang Xu, Ruirong Lin, Cen Jiang, and Xiangjin Chen
- Subjects
Epigenomics ,Male ,Antimetabolites, Antineoplastic ,Cancer Research ,PAX5 Transcription Factor ,Elongator complex ,Mice, Nude ,Decitabine ,Deoxycytidine ,DNA methyltransferase ,Mice ,chemistry.chemical_compound ,Transcription (biology) ,Animals ,Humans ,Epigenetics ,RC254-282 ,DNA methylation ,Chemistry ,Research ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Gallbladder cancer ,Gemcitabine ,Chromatin ,Demethylating agent ,Oncology ,CpG site ,Cancer research ,Gallbladder Neoplasms - Abstract
Background Gallbladder cancer (GBC) is known for its high malignancy and multidrug resistance. Previously, we uncovered that impaired integrity and stability of the elongator complex leads to GBC chemotherapy resistance, but whether its restoration can be an efficient therapeutic strategy for GBC remains unknown. Methods RT-qPCR, MS-qPCR and ChIP-qPCR were used to evaluate the direct association between ELP5 transcription and DNA methylation in tumour and non-tumour tissues of GBC. EMSA, chromatin accessibility assays, and luciferase assays were utilized to analysis the DNA methylation in interfering PAX5-DNA interactions. The functional experiments in vitro and in vivo were performed to investigate the effects of DNA demethylating agent decitabine (DAC) on the transcription activation of elongator complex and the enhanced sensitivity of gemcitabine in GBC cells. Tissue microarray contains GBC tumour tissues was used to evaluate the association between the expression of ELP5, DNMT3A and PAX5. Results We demonstrated that transcriptional repression of ELP5 in GBC was highly correlated with hypermethylation of the promoter. Mechanistically, epigenetic analysis revealed that DNA methyltransferase DNMT3A-catalysed hypermethylation blocked transcription factor PAX5 activation of ELP5 by disrupting PAX5-DNA interaction, resulting in repressed ELP5 transcription. Pharmacologically, the DNA demethylating agent DAC eliminated the hypermethylated CpG dinucleotides in the ELP5 promoter and then facilitated PAX5 binding and reactivated ELP5 transcription, leading to the enhanced function of the elongator complex. To target this mechanism, we employed a sequential combination therapy of DAC and gemcitabine to sensitize GBC cells to gemcitabine-therapy through epigenetic activation of the elongator complex. Conclusions Our findings suggest that ELP5 expression in GBC is controlled by DNA methylation-sensitive induction of PAX5. The sequential combination therapy of DAC and gemcitabine could be an efficient therapeutic strategy to overcome chemotherapy resistance in GBC.
- Published
- 2021
25. Does dihydropyrimidine dehydrogenase level modify plasma antioxidant capacity in colorectal cancer patients treated with fluoropyrimidines?
- Author
-
Marin P. Angelov, Velko Minchev, Kalina Kamenova, Liliya A. Atanasova, Slavina Surcheva, Nadya Hristova-Avakumova, Rumen Nikolov, and Lozan Todorov
- Subjects
Antimetabolites, Antineoplastic ,ABTS ,Colorectal cancer ,business.industry ,dihydropyrimidine dehydro ,colorectal cancer ,General Medicine ,Pharmacology ,medicine.disease ,Antioxidants ,Antioxidant capacity ,chemistry.chemical_compound ,chemistry ,medicine ,Dihydropyrimidine dehydrogenase ,Humans ,Medicine ,Fluorouracil ,Colorectal Neoplasms ,business ,Dihydrouracil Dehydrogenase (NADP) - Abstract
Introduction: Colorectal cancer is the third most common cancer type worldwide. Fluoropyrimidines and their prodrug-based regimens are widely applied as primary medications. The main enzyme responsible for the rate-limiting step in pyrimidine and for the 5-fluorouracil catabolism is dihydropyrimidine dehydrogenase (DPD). Aim: We aimed to screen DPD level and the changes of plasma antioxidant capacity of colorectal cancer patients on 5-fluorouracil regimen. Materials and methods: Human DPD Elisa Kit based on sandwich enzyme-linked immune-sorbent assay and spectrophotometric methods (FRAP and ABTS) were used in the study. Results: No statistically significant changes in plasma scavenging activity according to the results obtained in the ABTS system have been observed after evaluating all patients and considering DPD concentration. A decrease of the ferric reducing ability of patients’ plasma taken after the administered treatment was found. The increase of DPD level is accompanied by a decrease in the p values and therefore the statistical significance of the differences increases. Conclusions: Based on the aforementioned observations, it could be concluded that some aspects of plasma antioxidant capacity and individuals’ antioxidant status might be involved in the pathogenesis of the disease and could be altered by the activity of some enzymes. The cancer therapy in question, by the specificity of its mechanism of action, can modify patient’s oxidative status.
- Published
- 2021
26. Half-dose glucarpidase as efficient rescue for toxic methotrexate levels in patients with acute kidney injury
- Author
-
Raphael Teipel, Susanne Quick, Stephan Richter, Holger Knoth, Theresa Kretschmann, Frank Kroschinsky, Christoph Röllig, Ekaterina Balaian, Nael Alakel, Martin Bornhäuser, Sandra Heuschkel, Malte von Bonin, and Simone von Bonin
- Subjects
Adult ,Male ,Drug ,Antimetabolites, Antineoplastic ,Cancer Research ,media_common.quotation_subject ,Pharmacology ,Toxicology ,Folinic acid ,chemistry.chemical_compound ,medicine ,Humans ,Pharmacology (medical) ,In patient ,Adverse effect ,Aged ,media_common ,Creatinine ,Glucarpidase ,business.industry ,Acute kidney injury ,gamma-Glutamyl Hydrolase ,Acute Kidney Injury ,Middle Aged ,medicine.disease ,Thrombocytopenia ,Methotrexate plasma concentration ,Recombinant Proteins ,Methotrexate ,High-dose methotrexate ,Oncology ,chemistry ,Half-dose glucarpidase ,Female ,Original Article ,business ,medicine.drug - Abstract
Purpose High-dose methotrexate (HDMTX)-associated acute kidney injury with delayed MTX clearance has been linked to an excess in MTX-induced toxicities. Glucarpidase is a recombinant enzyme that rapidly hydrolyzes MTX into non-toxic metabolites. The recommended dose of glucarpidase is 50 U/kg, which has never been formally established in a dose finding study in humans. Few case reports, mostly in children, suggest that lower doses of glucarpidase might be equally effective in lowering MTX levels. Methods Seven patients with toxic MTX plasma concentrations following HDMTX therapy were treated with half-dose glucarpidase (mean 25 U/kg, range 17–32 U/kg). MTX levels were measured immunologically as well as by liquid chromatography–mass spectrometry (LC–MS). Toxicities were assessed according to National Cancer Institute—Common Terminology Criteria for Adverse Events (CTCAE) v5.0. Results All patients experienced HDMTX-associated kidney injury (median increase in creatinine levels within 48 h after HDMTX initiation compared to baseline of 251%, range 80–455%) and showed toxic MTX plasma concentrations (range 3.1–182.4 µmol/L) before glucarpidase injection. The drug was administered 42–70 h after HDMTX initiation. Within one day after glucarpidase injection, MTX plasma concentrations decreased by ≥ 97.7% translating into levels of 0.02–2.03 µmol/L. MTX rebound was detected in plasma 42–73 h after glucarpidase initiation, but concentrations remained consistent at Conclusion Half-dose glucarpidase seems to be effective in lowering MTX levels to concentrations manageable with continued intensified folinic acid rescue.
- Published
- 2021
27. Effectiveness and Safety of Clofarabine Monotherapy or Combination Treatment in Relapsed/Refractory Childhood Acute Lymphoblastic Leukemia: A Pragmatic, Non-interventional Study in Korea
- Author
-
Jung Woo Han, Jae Wook Lee, Hong Hoe Koo, Hana Cho, Che Ry Hong, Hyery Kim, Kyung-Nam Koh, Pil Sang Jang, Nack-Gyun Chung, Hoon Kook, Hee Young Shin, Bin Cho, Eu Jeen Yang, In Sang Jeon, Ho Joon Im, Keon Hee Yoo, Kyung Taek Hong, Chuhl Joo Lyu, Jung Yoon Choi, Young Tak Lim, Jong Jin Seo, Hyoung Jin Kang, Seongkoo Kim, and Seung Min Hahn
- Subjects
Adult ,Male ,0301 basic medicine ,Antimetabolites, Antineoplastic ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Combination therapy ,Pediatric Cancer ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Acute lymphoblastic leukemia ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Internal medicine ,Republic of Korea ,medicine ,Humans ,Clofarabine ,Prospective Studies ,Child ,Adverse effect ,Childhood Acute Lymphoblastic Leukemia ,Salvage Therapy ,Leukemia ,business.industry ,Hematopoietic Stem Cell Transplantation ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Prognosis ,Combined Modality Therapy ,Pediatric cancer ,Pediatric malignancy ,Survival Rate ,Regimen ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Child, Preschool ,030220 oncology & carcinogenesis ,Female ,Original Article ,Neoplasm Recurrence, Local ,business ,Follow-Up Studies ,medicine.drug - Abstract
Purpose Effectiveness and safety of clofarabine (one of the treatment mainstays in pediatric patients with relapsed/refractory acute lymphoblastic leukemia [ALL]) was assessed in Korean pediatric patients with ALL to facilitate conditional coverage with evidence development.Materials and Methods In this multicenter, prospective, observational study, patients receiving clofarabine as mono/combination therapy were followed up every 4-6 weeks for 6 months or until hematopoietic stem cell transplantation (HSCT). Response rates, survival outcomes, and adverse events were assessed.Results Sixty patients (2-26 years old; 65% B-cell ALL, received prior ≥ 2 regimen, 68.3% refractory to previous regimen) were enrolled and treated with at least one dose of clofarabine; of whom 26 (43.3%) completed 6 months of follow-up after the last dose of clofarabine. Fifty-eight patients (96.7%) received clofarabine combination therapy. Overall remission rate (complete remission [CR] or CR without platelet recovery [CRp]) was 45.0% (27/60; 95% confidence interval [CI], 32.4 to 57.6) and the overall response rate (CR, CRp, or partial remission [PR]) was 46.7% (28/60; 95% CI, 34.0 to 59.3), with 11 (18.3%), 16 (26.7%), and one (1.7%) patients achieving CR, CRp, and PR, respectively. The median time to remission was 5.1 weeks (95% CI, 4.7 to 6.1). Median duration of remission was 16.6 weeks (range, 2.0 to 167.6 weeks). Sixteen patients (26.7%) proceeded to HSCT. There were 24 deaths; 14 due to treatment-emergent adverse events.Conclusion Remission with clofarabine was observed in approximately half of the study patients who had overall expected safety profile; however, there was no favorable long-term survival outcome in this study.
- Published
- 2021
28. RING-finger protein 6 promotes colorectal tumorigenesis by transcriptionally activating SF3B2
- Author
-
Yunfei Zhou, Lei Liu, Weilin Li, Hui Xu, Jun Yu, Yan Li, Lifu Wang, and Chi Chun Wong
- Subjects
Male ,Antimetabolites, Antineoplastic ,Cancer Research ,Carcinogenesis ,Transgene ,Mice, Nude ,Apoptosis ,Biology ,Article ,Mice ,Gastrointestinal cancer ,Downregulation and upregulation ,In vivo ,Splicing factor 3b subunit 2 ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Genetics ,Organoid ,Animals ,Humans ,Chemotherapy ,Molecular Biology ,Cell Proliferation ,Mice, Inbred BALB C ,Cell growth ,Oncogenes ,Prognosis ,Xenograft Model Antitumor Assays ,In vitro ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Survival Rate ,Case-Control Studies ,Cancer research ,Epoxy Compounds ,Drug Therapy, Combination ,Fluorouracil ,Macrolides ,RNA Splicing Factors ,Colorectal Neoplasms ,Chromatin immunoprecipitation - Abstract
RNF6 is a RING finger protein with oncogenic potential. In this study, we established colon-specific RNF6 transgenic (tg) mice, and demonstrated that RNF6 overexpression accelerated colorectal carcinogenesis compared to wild-type littermates in a chemically induced colorectal cancer (CRC) model. To understand whether transcriptional activity of RNF6 underlies its oncogenic effect, we performed integrated chromatin immunoprecipitation (ChIP)-sequencing and RNA-sequencing analysis to identify splicing factor 3b subunit 2 (SF3B2) as a potential downstream target of RNF6. RNF6 binds to the SF3B2 promoter and the overexpression of RNF6 activates SF3B2 expression in CRC cells, primary CRC organoids, and RNF6 tg mice. SF3B2 knockout abrogated the tumor promoting effect of RNF6 overexpression, whereas the reexpression of SF3B2 recused cell growth and migration/invasion in RNF6 knockout cells, indicating that SF3B2 is a functional downstream target of RNF6 in CRC. Targeting of RNF6-SF3B2 axis with SF3B2 inhibitor with pladienolide B suppressed the growth of CRC cells with RNF6 overexpression in vitro and in vivo. Moreover, the combination of 5-fluorouracil (5-FU) plus pladienolide B exerted synergistic effects in CRC with high RNF6 expression, leading to tumor regression in xenograft models. These findings indicate that tumor promoting effect of RNF6 is achieved mainly via transcriptional upregulation of SF3B2, and that RNF6-SF3B2 axis is a promising target for CRC therapy.
- Published
- 2021
29. Exosomal lncRNA FOXD3-AS1 upregulates ELAVL1 expression and activates PI3K/Akt pathway to enhance lung cancer cell proliferation, invasion, and 5-fluorouracil resistance
- Author
-
Guangxian Mao, Zhimin Mu, and Da Wu
- Subjects
Male ,Antimetabolites, Antineoplastic ,Lung Neoplasms ,animal structures ,Biophysics ,Adenocarcinoma of Lung ,Exosomes ,Biochemistry ,ELAV-Like Protein 1 ,Phosphatidylinositol 3-Kinases ,Cell Movement ,Cell Line, Tumor ,medicine ,Humans ,Neoplasm Invasiveness ,RNA, Small Interfering ,Lung cancer ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,A549 cell ,Phosphoinositide 3-kinase ,biology ,Akt/PKB signaling pathway ,Cell growth ,Forkhead Transcription Factors ,General Medicine ,Transfection ,Middle Aged ,respiratory system ,medicine.disease ,Small Cell Lung Carcinoma ,respiratory tract diseases ,Gene Expression Regulation, Neoplastic ,A549 Cells ,Drug Resistance, Neoplasm ,embryonic structures ,Carcinoma, Squamous Cell ,biology.protein ,Cancer research ,Female ,RNA, Long Noncoding ,Fluorouracil ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Long non-coding RNA (lncRNA) FOXD3-AS1 expression is upregulated in lung cancer; however, its effect and mechanism on 5-fluorouracil (5-FU) resistance remain unclear. In this study, we determined the effects of FOXD3-AS1-enriched exosomes derived from lung cancer cells on the proliferation, invasion, and 5-FU resistance of lung cancer cells. Online bioinformatics database analysis showed that FOXD3-AS1 was upregulated in lung cancer progression. Real-time quantitative PCR results confirmed that FOXD3-AS1 expression was upregulated in lung cancer tissues and cell lines, and FOXD3-AS1 was greatly enriched in lung cancer cell-derived exosomes. ELAV-like RNA-binding protein 1 (ELAVL1) was identified as an RNA-binding protein of FOXD3-AS1. The lung cancer cell-derived exosomes promoted A549 cell proliferation and invasion and inhibited apoptosis caused by 5-FU, and transfection of si-FOXD3-AS1 or si-ELAVL1 in exosome-incubated A549 cells reversed these effects. Moreover, exosome-incubated A549 cells were co-transfected with si-FOXD3-AS1 and pcDNA-ELAVL1, showing the same cell proliferation, invasion, and 5-FU resistance as those of A549 cells treated with lung cancer cell-derived exosomes alone. Mechanistic studies identified that lung cancer cell-derived exosomes activated the PI3K/Akt pathway, and transfection of si-FOXD3-AS1 or treatment with the PI3K inhibitor LY294002 reversed the activation of the PI3K/Akt axis induced by exosomes. In conclusion, our study revealed that lung cancer cell-derived exosomal FOXD3-AS1 upregulated ELAVL1 expression and activated the PI3K/Akt pathway to promote lung cancer progression. Our findings provide a new strategy for lung cancer treatment.
- Published
- 2021
30. 2’-Fucosyllactose Ameliorates Chemotherapy-Induced Intestinal Mucositis by Protecting Intestinal Epithelial Cells Against Apoptosis
- Author
-
Steven D. Townsend, Yaohua Yang, Jirong Long, Fang Yan, M. Kay Washington, Gang Zhao, and Jessica Williams
- Subjects
Proliferation ,MSIE, mouse small intestine epithelial cells ,MPO, myeloperoxidase ,Apoptosis ,RC799-869 ,IEC, intestinal epithelial cell ,Mice ,PCR, polymerase chain reaction ,Human Milk Oligosaccharides ,Original Research ,TNF, tumor necrosis factor ,Gastroenterology ,ELISA, enzyme-linked immunosorbent assay ,Diseases of the digestive system. Gastroenterology ,2’-FL, 2’-fucosyllactose ,mRNA, messenger RNA ,medicine.anatomical_structure ,LPS, lipopolysaccharide ,FITC, fluorescein isothiocyanate ,Fluorouracil ,Goblet Cells ,medicine.symptom ,Diarrhea ,Mucositis ,Antimetabolites, Antineoplastic ,5-Fluorouracil ,EdU, 5-ethynyl-2'-deoxyuridine ,PBS, phosphate-buffered saline ,Inflammation ,Proinflammatory cytokine ,HT29 Cells ,FBS, fetal bovine serum ,medicine ,Animals ,HMO, human milk oligosaccharide ,NMR, nuclear magnetic resonance ,SV40, simian virus 40 ,ZO-1, Zona occludin-1 ,Goblet cell ,Hepatology ,Cell growth ,business.industry ,medicine.disease ,Small intestine ,IL, interleukin ,Cancer research ,5-FU, 5-fluorouracil ,business ,Trisaccharides ,Intestinal Inflammation - Abstract
Background & Aims Intestinal mucositis, a severe complication of antineoplastic therapeutics, is characterized by mucosal injury and inflammation in the small intestine. Therapies for the prevention and treatment of this disease are needed. We investigated whether 2’-fucosyllactose (2’-FL), an abundant oligosaccharide in human milk, protects intestinal integrity and ameliorates intestinal mucositis. Methods A mouse small intestinal epithelial (MSIE) cell line, mouse enteroid cultures, and human gastrointestinal tumor cell lines (AGS and HT29) were co-treated with the chemotherapy agent 5-fluorouracil (5-FU) and 2’-FL. Mice were injected intraperitoneally with 5-FU to induce intestinal mucositis. 2’-FL was administered in the drinking water to mice before (pretreatment) or concurrently with 5-FU injection. Body weight and pathologic changes were analyzed. Results 2’-FL alleviated 5-FU inhibition of cell growth in MSIE cells, but not in AGS and HT29 cells. The 5-FU–induced apoptosis in MSIE cells and enteroids was suppressed by 2’-FL. Compared with 5-FU treatment alone, 2’-FL pretreatment protected against body weight loss, and ameliorated inflammation scores, proinflammatory cytokine production, shortening of villi, epithelial cell apoptosis, goblet cell loss, and tight junctional complex disruption in the small intestine. 2’-FL concurrent treatment had less of an effect on intestinal mucositis than 2’-FL pretreatment. Interestingly, no effect of 2’-FL was observed on 5-FU–induced S-phase arrest in MSIE, AGS, and HT29 cells. Neither pretreatment nor concurrent treatment with 2’-FL affected 5-FU–induced inhibition of proliferation in MSIE cells. Conclusions This study shows a novel direct effect of 2’-FL in protecting small intestinal epithelial cells against apoptosis stimulated by 5-FU, which may contribute to prevention of 5-FU–induced intestinal mucositis., Graphical abstract
- Published
- 2021
31. Mutual Prodrugs of 5‐Fluorouracil: From a Classic Chemotherapeutic Agent to Novel Potential Anticancer Drugs
- Author
-
Loredana Salerno, Valeria Ciaffaglione, Giuseppe Romeo, Sebastiano Intagliata, Maria N. Modica, and Valeria Pittalà
- Subjects
Antimetabolites, Antineoplastic ,5-Fluorouracil ,Organizational model ,Context (language use) ,Drug resistance ,Bioinformatics ,Biochemistry ,World health ,Essential medicines ,Broad spectrum ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Humans ,Prodrugs ,5-FU hybrids ,Enzyme Inhibitors ,General Pharmacology, Toxicology and Pharmaceutics ,Pharmacology ,5-FU conjugates ,business.industry ,Organic Chemistry ,Drug Synergism ,Prodrug ,Fluorouracil ,Anticancer agents ,Molecular Medicine ,business ,Mutual prodrugs ,medicine.drug - Abstract
The development of potent antitumor agents with a low toxicological profile against healthy cells is still one of the greatest challenges facing in medicinal chemistry. In this context, the "mutual prodrug" approach has emerged as a potential tool to overcome undesirable physicochemical features and mitigate the side effects of approved drugs. Among broad spectrum chemotherapeutics available for clinical use today, 5-fluorouracil (5-FU) is one of the most representative, also included in the World Health Organization model list of essential medicines. Unfortunately, severe side effects and drug resistance phenomena are still the primary limits and drawbacks in its clinical use. This review describes the progress made over the last ten years in developing 5-FU-based mutual prodrugs to improve the therapeutic profile and achieve targeted delivery to cancer tissues.
- Published
- 2021
32. A Modified Regimen of 21-day Nab-Paclitaxel Plus Gemcitabine in Locally Advanced or Metastatic Pancreatic Cancer: A Retrospective Real-World Study
- Author
-
Chen Chang, Lingwei Meng, Xiaofen Li, Ke Cheng, Cheng Yi, Bing Peng, Ji Ma, and Dan Cao
- Subjects
Pancreatic Neoplasms ,Antimetabolites, Antineoplastic ,Oncology ,Paclitaxel ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Hematology ,General Medicine ,Gemcitabine ,Retrospective Studies - Abstract
Background It is 1 of the standard treatment options for metastasis pancreatic cancer to receive nab-paclitaxel (125 mg/m2) plus gemcitabine (1000 mg/m2) on days 1, 8 and 15 every 28 days. Some patients showed intolerance and inconvenience to this therapeutic regimen. Thus, we conducted this retrospective real-world study to determine the efficacy and tolerability of a modified 21-day nab-paclitaxel plus gemcitabine (nab-P/Gem) regimen for the first-line treatment of locally advanced or metastatic pancreatic cancer. Methods Patients with locally advanced and metastatic pancreatic cancer treated with nab-paclitaxel (125 mg/m2) plus gemcitabine (1000 mg/m2) on days 1 and 8 every 21-day at West China Hospital and Shang Jin Hospital of Sichuan University from Mar 2018 to Dec 2021 were reviewed retrospectively. Clinical characteristics of patients were collected. The progression-free survival, overall survival, objective response rate, disease control rate, and toxicity were evaluated. Results A total of 113 patients who received the modified regimen of 21-day nab-P/Gem chemotherapy were included. The median overall survival was 9.3 months and the median progression-free survival was 4.4 months. The objective response rate and disease control rate were 18.6% and 56.7%, respectively. The median relative dose intensity for this modified regimen was 65%. The adverse events were mild to moderate, and the most common grade 3 or 4 treatment-related adverse events were neutropenia (21%) and leukopenia (16%). Conclusions Our study showed that this modified regimen of 21-day nab-P/Gem for locally advanced and metastatic pancreatic cancer had comparable efficacy and tolerable toxicity. This treatment may provide a considerable option for pancreatic cancer patients who desire a modified schedule. The modified regimen of 21-day nab-P/Gem is also an option worth considering during the coronavirus disease 2019 pandemic for minimizing the number of visits and limiting the risk of exposure.
- Published
- 2022
33. Predicting Dihydropyrimidine Dehydrogenase Deficiency and Related 5-Fluorouracil Toxicity: Opportunities and Challenges of
- Author
-
Ottavia, De Luca, Gerardo, Salerno, Donatella, De Bernardini, Maria Simona, Torre, Maurizio, Simmaco, Luana, Lionetto, Giovanna, Gentile, and Marina, Borro
- Subjects
Antimetabolites, Antineoplastic ,Dihydropyrimidine Dehydrogenase Deficiency ,Humans ,Fluorouracil ,Exons ,Floxuridine ,Dihydrouracil Dehydrogenase (NADP) - Abstract
Deficiency of dihydropyrimidine dehydrogenase (DPD), encoded by the
- Published
- 2022
34. The Co‐mutational Spectrum Determines the Therapeutic Response in Murine FGFR2 Fusion‐Driven Cholangiocarcinoma
- Author
-
Jens U. Marquardt, Michael Saborowski, Silke Marhenke, Ralph M. Wirtz, Andreas Pich, Arndt Vogel, Anna Saborowski, Gajanan Kendre, Norman Woller, Tanja Poth, D Becker, Karthikeyan Murugesan, Tanja Reineke-Plaaß, Georgina Lorz, and Florian Kühnel
- Subjects
Fetal Proteins ,0301 basic medicine ,Antimetabolites, Antineoplastic ,Combination therapy ,medicine.medical_treatment ,FGFR Inhibition ,Vesicular Transport Proteins ,Cyclic AMP Response Element-Binding Protein A ,medicine.disease_cause ,Deoxycytidine ,Malignant transformation ,Targeted therapy ,Cholangiocarcinoma ,Proto-Oncogene Proteins p21(ras) ,Mice ,03 medical and health sciences ,Liver Neoplasms, Experimental ,0302 clinical medicine ,Antigens, Neoplasm ,medicine ,Animals ,Receptor, Fibroblast Growth Factor, Type 2 ,Protein Kinase Inhibitors ,Cell Proliferation ,Hepatology ,Oncogene ,business.industry ,Fibroblast growth factor receptor 2 ,Adenosylhomocysteinase ,Phenylurea Compounds ,Gemcitabine ,Bile Ducts, Intrahepatic ,Cell Transformation, Neoplastic ,Pyrimidines ,030104 developmental biology ,Bile Duct Neoplasms ,Fibroblast growth factor receptor ,Mutation ,Cancer research ,030211 gastroenterology & hepatology ,KRAS ,Gene Fusion ,business ,Co-Repressor Proteins ,Microtubule-Associated Proteins - Abstract
Background and aims Intrahepatic cholangiocarcinoma (ICC) is the second most common primary liver cancer and a highly lethal malignancy. Chemotherapeutic options are limited, but a considerable subset of patients harbors genetic lesions for which targeted agents exist. Fibroblast growth factor receptor 2 (FGFR2) fusions belong to the most frequent and therapeutically relevant alterations in ICC, and the first FGFR inhibitor was recently approved for the treatment of patients with progressed, fusion-positive ICC. Response rates of up to 35% indicate that FGFR-targeted therapies are beneficial in many but not all patients. Thus far, no established biomarkers exist that predict resistance or response to FGFR-targeted therapies in patients with ICC. Approach and results In this study, we use an autochthonous murine model of ICC to demonstrate that FGFR2 fusions are potent drivers of malignant transformation. Furthermore, we provide preclinical evidence that the co-mutational spectrum acts not only as an accelerator of tumor development, but also modifies the response to targeted FGFR inhibitors. Using pharmacologic approaches and RNA-interference technology, we delineate that Kirsten rat sarcoma oncogene (KRAS)-activated mitogen-activated protein kinase signaling causes primary resistance to FGFR inhibitors in FGFR2 fusion-positive ICC. The translational relevance is supported by the observation that a subset of human FGFR2 fusion patients exhibits transcriptome profiles reminiscent of KRAS mutant ICC. Moreover, we demonstrate that combination therapy has the potential to overcome primary resistance and to sensitize tumors to FGFR inhibition. Conclusions Our work highlights the importance of the co-mutational spectrum as a significant modifier of response in tumors that harbor potent oncogenic drivers. A better understanding of the genetic underpinnings of resistance will be pivotal to improve biomarker-guided patient selection and to design clinically relevant combination strategies.
- Published
- 2021
35. Puerarin Ameliorates 5-Fluorouracil–Induced Intestinal Mucositis in Mice by Inhibiting JAKs
- Author
-
Jingyu Wang, Dapeng Chen, Shuaishuai Zhang, Yan Hu, Liang Wang, Baohui Song, and Jun Chen
- Subjects
Male ,Mucositis ,Antimetabolites, Antineoplastic ,Vasodilator Agents ,medicine.medical_treatment ,Intraperitoneal injection ,Inflammation ,Pharmacology ,Proinflammatory cytokine ,Mice ,chemistry.chemical_compound ,Puerarin ,medicine ,Animals ,Humans ,Janus Kinase Inhibitors ,Intestinal Mucosa ,Chemotherapy ,Dose-Response Relationship, Drug ,business.industry ,medicine.disease ,Isoflavones ,Rats ,Mice, Inbred C57BL ,chemistry ,Apoptosis ,Molecular Medicine ,Fluorouracil ,Caco-2 Cells ,medicine.symptom ,Janus kinase ,business - Abstract
Intestinal mucositis, resulting from 5-fluorouracil (5-FU)-based chemotherapy, subjects patients to great pain and hampers cancer treatment progress. Puerarin, the major active ingredient in Pueraria lobata, exerts anti-inflammatory and anti-oxidative effects. However, whether puerarin has an effect on 5-FU-induced intestinal mucositis remains unknown. We established a mice model of intestinal mucositis through the intraperitoneal injection of 5-FU, and then injected puerarin (50 and 100 mg/kg) intraperitoneally for 7 consecutive days. Routine parameters, such as body weight, food intake, and diarrheal incidence, were examined to evaluate the effects of puerarin on intestinal mucositis in mice. The intestinal barrier9s functions were also evaluated by measuring the serum recovery of fluorescein isothiocyanate-4kD dextran in this study. The expression levels of inflammatory cytokines, inflammatory mediators, oxidative reactions, as well as apoptotic marker proteins, were determined to elucidate the underlying mechanisms of puerarin on intestinal mucositis. The model mice presented symptoms and histopathological changes typical of 5-FU-induced intestinal mucositis. In addition to vigorous inflammatory reactions, oxidative reactions and cell apoptosis, Janus kinase (JAK) was markedly activated. Puerarin decreased the expression levels of those of inflammatory mediators, oxidative reactions, and apoptosis-related proteins in 5-FU-induced mucositis by blocking the activation of JAK. Puerarin decreased inflammation, oxidative reactions and apoptosis, and protected intestinal barrier functions, to ameliorate 5-FU-induced intestinal mucositis by inhibiting the activation of JAK. This study provides novel insights into the pathological mechanisms of, and treatment alternatives for, 5-FU-induced intestinal mucositis. Significance Statement This study reveals the mechanism responsible for the protective effects of puerarin in 5-fluorouracil-induced intestinal mucositis. Puerarin inhibits the activation of JAK, thereby suppressing inflammation, oxidative reactions, cell apoptosis, and protected intestinal barrier functions, to ameliorate 5-FU-induced intestinal mucositis. Overall, our results suggest that puerarin can serve as a potential natural JAK inhibitor in the treatment of 5-FU-induced intestinal mucositis.
- Published
- 2021
36. Current progress and future perspectives of research on intravascular large B‐cell lymphoma
- Author
-
Kazuyuki Shimada and Hitoshi Kiyoi
- Subjects
Male ,Oncology ,Cancer Research ,intravascular large B‐cell lymphoma ,Review Article ,Disease ,B7-H1 Antigen ,Central Nervous System Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Cumulative incidence ,Review Articles ,Injections, Spinal ,medicine.diagnostic_test ,biology ,General Medicine ,Middle Aged ,Progression-Free Survival ,Vascular Neoplasms ,Vincristine ,central nervous system involvement ,Heterografts ,Female ,Rituximab ,Lymphoma, Large B-Cell, Diffuse ,medicine.drug ,Antimetabolites, Antineoplastic ,medicine.medical_specialty ,Clinical Trials, Phase II as Topic ,Internal medicine ,PD-L1 ,Biopsy ,medicine ,Humans ,Cyclophosphamide ,Intravascular large B-cell lymphoma ,genetic alterations ,business.industry ,Cell Cycle Checkpoints ,Programmed Cell Death 1 Ligand 2 Protein ,medicine.disease ,Lymphoma ,Methotrexate ,Doxorubicin ,PD‐L1 ,biology.protein ,Prednisone ,R‐CHOP therapy ,business ,Neoplasm Transplantation ,Forecasting ,Rare disease - Abstract
Intravascular large B‐cell lymphoma is a rare disease of the large B cells characterized by selective growth in the lumina of small vessels in systemic organs. Since first reported in 1959, the difficulty of obtaining sufficient tumor cells from biopsy specimens has hampered the elucidation of its underlying biology. Recent progress using xenograft models and plasma cell‐free DNA has uncovered genetic features that are similar to those of activated B‐cell type diffuse large B‐cell lymphoma, including MYD88 and CD79B mutations and frequent alterations in immune check point‐related genes such as PD‐L1 and PD‐L2. Given the improvement in clinical outcomes and a higher risk of secondary central nervous system (CNS) involvement in the rituximab era, a phase 2 trial of R‐CHOP combined with high‐dose methotrexate and intrathecal chemotherapy as a CNS‐oriented therapy has been conducted. This trial, the PRIMEUR‐IVL study, has displayed good progression‐free survival and a low cumulative incidence of secondary CNS involvement. Long‐term follow‐up within this trial is still ongoing. Further understanding of the pathophysiology of the disease and improvements in clinical outcomes are still needed., Intravascular large B‐cell lymphoma is a rare type of extranodal large B‐cell lymphoma. Recent progress on clinical and translational research on IVLBCL allows us to understand the biological features of the disease and to apply an active treatment. Further investigations are warranted to deepen our understanding of the disease and to establish optimal treatments.
- Published
- 2021
37. New insights into the mechanisms underlying 5-fluorouracil-induced intestinal toxicity based on transcriptomic and metabolomic responses in human intestinal organoids
- Author
-
Johanna Vappiani, Terezinha de Souza, Theo M. de Kok, Luke Coyle, Julio Saez-Rodriguez, Danyel Jennen, Daniel C. Sévin, D. F. Rodrigues, Seung-Wook Chung, Matteo Di Piazza, Bram Herpers, Attila Gábor, Mian Zhang, Anthony M. Lynch, Jos C. S. Kleinjans, Sofia Ferreira, Toxicogenomics, RS: GROW - R1 - Prevention, RS: FSE MaCSBio, RS: FPN MaCSBio, and RS: FHML MaCSBio
- Subjects
0301 basic medicine ,Male ,PHARMACOKINETICS ,Health, Toxicology and Mutagenesis ,Cell ,Apoptosis ,Toxicology ,Toxicogenomics and Omics Technologies ,COLORECTAL-CANCER ,Transcriptome ,0302 clinical medicine ,Intestine, Small ,E2F1 ,TISSUE DISTRIBUTION ,GENE-EXPRESSION ,Cell Cycle ,General Medicine ,Cell cycle ,STEM ,5-FU toxicity ,3. Good health ,Cell biology ,Organoids ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Antimetabolites, Antineoplastic ,Cell Survival ,Colon ,Human organoid models ,Biology ,HIGH-THROUGHPUT ,Models, Biological ,03 medical and health sciences ,Metabolomics ,Organoid ,Metabolome ,medicine ,Humans ,Transcriptomics ,MUCOSAL INJURY ,Transcription factor ,Dose-Response Relationship, Drug ,Molecular mechanisms ,IN-VITRO ,FLUOROURACIL ,MODEL ,Oxidative Stress ,030104 developmental biology - Abstract
5-Fluorouracil (5-FU) is a widely used chemotherapeutical that induces acute toxicity in the small and large intestine of patients. Symptoms can be severe and lead to the interruption of cancer treatments. However, there is limited understanding of the molecular mechanisms underlying 5-FU-induced intestinal toxicity. In this study, well-established 3D organoid models of human colon and small intestine (SI) were used to characterize 5-FU transcriptomic and metabolomic responses. Clinically relevant 5-FU concentrations for in vitro testing in organoids were established using physiologically based pharmacokinetic simulation of dosing regimens recommended for cancer patients, resulting in exposures to 10, 100 and 1000 µM. After treatment, different measurements were performed: cell viability and apoptosis; image analysis of cell morphological changes; RNA sequencing; and metabolome analysis of supernatant from organoids cultures. Based on analysis of the differentially expressed genes, the most prominent molecular pathways affected by 5-FU included cell cycle, p53 signalling, mitochondrial ATP synthesis and apoptosis. Short time-series expression miner demonstrated tissue-specific mechanisms affected by 5-FU, namely biosynthesis and transport of small molecules, and mRNA translation for colon; cell signalling mediated by Rho GTPases and fork-head box transcription factors for SI. Metabolomic analysis showed that in addition to the effects on TCA cycle and oxidative stress in both organoids, tissue-specific metabolic alterations were also induced by 5-FU. Multi-omics integration identified transcription factor E2F1, a regulator of cell cycle and apoptosis, as the best key node across all samples. These results provide new insights into 5-FU toxicity mechanisms and underline the relevance of human organoid models in the safety assessment in drug development. Supplementary Information The online version contains supplementary material available at 10.1007/s00204-021-03092-2.
- Published
- 2021
38. Management of adverse events in patients with acute myeloid leukemia in remission receiving oral azacitidine: experience from the phase 3 randomized QUAZAR AML-001 trial
- Author
-
Jianhua Zhong, Hamid Sayar, Dominik Selleslag, Christopher Pocock, C.L. Beach, Farhad Ravandi, Rochelle Bailey, Pau Montesinos, Angela Figuera-Alvarez, Sang Kyun Sohn, Andrew H. Wei, Hervé Dombret, William Tse, Hartmut Döhner, Francesca Pierdomenico, Maurizio Musso, Gail J. Roboz, Timothy Chevassut, Hana Safah, Devendra K Hiwase, Ignazia La Torre, and Barry S. Skikne
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,Neutropenia ,Gastrointestinal Diseases ,Maintenance ,Phases of clinical research ,Administration, Oral ,Placebo ,Oral Azacitidine ,Maintenance therapy ,Internal medicine ,medicine ,Humans ,Diseases of the blood and blood-forming organs ,Adverse effect ,Molecular Biology ,RC254-282 ,Aged ,Aged, 80 and over ,Oral azacitidine ,business.industry ,Myelodysplastic syndromes ,Research ,Remission Induction ,Disease Management ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Anemia ,Hematology ,Middle Aged ,medicine.disease ,Placebo Effect ,Thrombocytopenia ,Discontinuation ,Leukemia, Myeloid, Acute ,Oncology ,Concomitant ,Azacitidine ,Female ,Safety ,RC633-647.5 ,business ,CC-486 - Abstract
Background Most older patients with acute myeloid leukemia (AML) who attain morphologic remission with intensive chemotherapy (IC) will eventually relapse and post-relapse prognosis is dismal. In the pivotal QUAZAR AML-001 trial, oral azacitidine maintenance therapy significantly prolonged overall survival by 9.9 months (P P Methods QUAZAR AML-001 is an international, placebo-controlled randomized phase 3 study. Patients aged ≥ 55 years with AML and intermediate- or poor-risk cytogenetics at diagnosis, who had attained first complete remission (CR) or CR with incomplete blood count recovery (CRi) within 4 months before study entry, were randomized 1:1 to receive oral azacitidine 300 mg or placebo once-daily for 14 days in repeated 28-day cycles. Safety was assessed in all patients who received ≥ 1 dose of study drug. Results A total of 469 patients received oral azacitidine (n = 236) or placebo (n = 233). Median age was 68 years. Patients received a median of 12 (range 1–80) oral azacitidine treatment cycles or 6 (1–73) placebo cycles. Gastrointestinal AEs were common and typically low-grade. The most frequent grade 3–4 AEs during oral azacitidine therapy were hematologic events. AEs infrequently required permanent discontinuation of oral azacitidine (13%), suggesting they were effectively managed with use of concomitant medications and oral azacitidine dosing modifications. Conclusion Oral azacitidine maintenance had a generally favorable safety profile. Prophylaxis with antiemetic agents, and blood count monitoring every other week, are recommended for at least the first 2 oral azacitidine treatment cycles, and as needed thereafter. Awareness of the type, onset, and duration of common AEs, and implementation of effective AE management, may maximize treatment adherence and optimize the survival benefits of oral azacitidine AML remission maintenance therapy. Trial registration This trial is registered on clinicaltrials.gov: NCT01757535 as of December 2012.
- Published
- 2021
39. TET2 Mutation and High miR-22 Expression as Biomarkers to Predict Clinical Outcome in Myelodysplastic Syndrome Patients Treated with Hypomethylating Therapy
- Author
-
Sung Hee Lim, Chan Kyu Kim, Jina Yun, Se Hyung Kim, Jong Ho Won, Seong Kyu Park, Sang Byung Bae, Young Sok Ji, and Geum Ha Jang
- Subjects
Microbiology (medical) ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,QH301-705.5 ,miR-22 ,Disease ,Decitabine ,Microbiology ,Dioxygenases ,hypomethylating therapy ,Young Adult ,Internal medicine ,Proto-Oncogene Proteins ,microRNA ,medicine ,Humans ,Biology (General) ,Molecular Biology ,Aged ,Aged, 80 and over ,TET2 ,business.industry ,Hazard ratio ,Tet methylcytosine dioxygenase 2 ,myelodysplastic syndrome (MDS) ,cytogenetic abnormality ,General Medicine ,Odds ratio ,DNA Methylation ,Middle Aged ,DNA-Binding Proteins ,Survival Rate ,hypermethylation ,MicroRNAs ,Treatment Outcome ,Myelodysplastic Syndromes ,Mutation (genetic algorithm) ,DNA methylation ,Mutation ,Azacitidine ,Biomarker (medicine) ,Female ,business ,Biomarkers - Abstract
Tet methylcytosine dioxygenase 2 (TET2) is one of the most frequently mutated genes in myelodysplastic syndrome (MDS). TET2 is known to involve a demethylation process, and the loss of TET2 is thought to cause DNA hypermethylation. Loss of TET2 function is known to be caused by genetic mutations and miRNA, such as miR-22. We analyzed 41 MDS patients receiving hypomethylating therapy (HMT) to assess whether TET2 mutation status and miR-22 expression status were associated with their clinical characteristics and treatment outcomes. Responsiveness to HMT was not affected by both TET2 mutation (odds ratio (OR) 0.900, p = 0.909) and high miR-22 expression (OR 1.548, p = 0.631). There was a tendency for TET2 mutation to be associated with lower-risk disease based on IPSS (Gamma = −0.674, p = 0.073), lower leukemic transformation (OR 0.170, p = 0.040) and longer survival (Hazard ratio 0.354, p = 0.059). Although high miR-22 expression also showed a similar tendency, this tendency was weaker than that of TET2 mutation. In summary, the loss of TET2 function, including both TET2 mutation and high miR-22 expression, was not a good biomarker for predicting the response to HMT but may be associated with lower-risk disease based on IPSS, lower leukemic transformation and longer survival.
- Published
- 2021
40. NKG2D defines tumor‐reacting effector CD8 + T cells within tumor microenvironment
- Author
-
Marija Mojic, Chikako Ohshima, Sisca Ucche, Yoshihiro Hayakawa, Kiyomi Shitaoka, Hideaki Tahara, Fulian Lyu, Hiroyuki Kishi, and Hiroshi Hamana
- Subjects
Antimetabolites, Antineoplastic ,Cancer Research ,Skin Neoplasms ,Ovalbumin ,medicine.medical_treatment ,Melanoma, Experimental ,CD8-Positive T-Lymphocytes ,NKG2D ,Interferon-gamma ,Mice ,Basic and Clinical Immunology ,Lymphocytes, Tumor-Infiltrating ,Antigens, Neoplasm ,Tumor Microenvironment ,medicine ,Animals ,Bioluminescence imaging ,Cytotoxic T cell ,Luciferases ,Mice, Knockout ,Tumor microenvironment ,Chemistry ,immune surveillance ,Vaccination ,Original Articles ,General Medicine ,Immunotherapy ,Tumor antigen ,TOX ,Mice, Inbred C57BL ,Bromodeoxyuridine ,Oncology ,NK Cell Lectin-Like Receptor Subfamily K ,Tumor progression ,Luminescent Measurements ,Cancer research ,Original Article ,CD8 ,cytotoxic T cell - Abstract
For successful immunotherapy for cancer, it is important to understand the immunological status of tumor antigen‐specific CD8+ T cells in the tumor microenvironment during tumor progression. In this study, we monitored the behavior of B16OVA‐Luc cells in mice immunized with a model tumor antigen ovalbumin (OVA). Using bioluminescence imaging, we identified the time series of OVA‐specific CD8+ T‐cell responses during tumor progression: initial progression, immune control, and the escape phase. As a result of analyzing the status of tumor antigen‐specific CD8+ cells in those 3 different phases, we found that the expression of NKG2D defines tumor‐reacting effector CD8+ T cells. NKG2D may control the fate and TOX expression of tumor‐reacting CD8+ T cells, considering that NKG2D blockade in OVA‐vaccinated mice delayed the growth of the B16OVA‐Luc2 tumor and increased the presence of tumor‐infiltrating OVA‐specific CD8+ T cells., In this study, we identified the time series of OVA‐specific CD8+ T‐cell responses during tumor progression: initial progression, immune control, and the escape phase. As a result of analyzing the status of tumor antigen‐specific CD8+ cells in those 3 different phases, we found that the expression of NKG2D defines tumor‐reacting effector CD8+ T cells and may control the fate and TOX expression of tumor‐reacting CD8 T cells.
- Published
- 2021
41. Myelodysplastic syndromes with 20q deletion: incidence, prognostic value and impact on response to azacitidine of ASXL1 chromosomal deletion and genetic mutations
- Author
-
Itziar Oiartzabal, Teresa Bernal, Carlos Solano, Eva Villamón, Míriam Gutiérrez, Rosario Abellán, Míriam Vara, María Díez-Campelo, María José Calasanz, Iván Martín, Sara Alvarez, Marisa Calabuig, Aroa Irigoyen, Guillermo Sanz, Isabel Granada, Esperanza Such, Elvira Mora, Rocío García‐Serra, Mª Laura Blanco, Rosa Collado, Rosana Diez, Andres Jerez, Blanca Xicoy, Angela Gil, and Mar Tormo
- Subjects
Male ,Oncology ,Antimetabolites, Antineoplastic ,azacitidine ,medicine.medical_specialty ,Azacitidine ,ASXL1 ,Gene mutation ,Internal medicine ,medicine ,Humans ,Gene ,Chromosomal Deletion ,Aged ,20q deletion ,gene mutations ,Aged, 80 and over ,business.industry ,Incidence ,Myelodysplastic syndromes ,Incidence (epidemiology) ,Hazard ratio ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,myelodysplastic syndromes ,Confidence interval ,Repressor Proteins ,Myelodysplastic Syndromes ,Mutation ,Female ,Chromosome Deletion ,business ,medicine.drug - Abstract
In myelodysplastic syndromes (MDS), the 20q deletion [del(20q)] may cause deletion of the ASXL1 gene. We studied 153 patients with MDS and del(20q) to assess the incidence, prognostic value and impact on response to azacitidine (AZA) of ASXL1 chromosomal alterations and genetic mutations. Additionally, in vitro assay of the response to AZA in HAP1 (HAP1(WT)) and HAP1 ASXL1 knockout (HAP1(KN)) cells was performed. ASXL1 chromosomal alterations were detected in 44 patients (28 center dot 5%): 34 patients (22%) with a gene deletion (ASXL1(DEL)) and 10 patients (6 center dot 5%) with additional gene copies. ASXL1(DEL) was associated with a lower platelet count. The most frequently mutated genes were U2AF1 (16%), ASXL1 (14%), SF3B1 (11%), TP53 (7%) and SRSF2 (6%). ASXL1 alteration due to chromosomal deletion or genetic mutation (ASXL1(DEL)/ASXL1(MUT)) was linked by multivariable analysis with shorter overall survival [hazard ratio, (HR) 1 center dot 84; 95% confidence interval, (CI): 1 center dot 11-3 center dot 04; P = 0 center dot 018] and a higher rate for acute myeloid leukaemia progression (HR 2 center dot 47; 95% CI: 1 center dot 07-5 center dot 70, P = 0 center dot 034). ASXL1(DEL)/ASXL1(MUT) patients were correlated by univariable analysis with a worse response to AZA. HAP1(KN) cells showed more resistance to AZA compared to HAP1(WT) cells. In conclusion, ASXL1 alteration exerts a negative impact on MDS with del(20q) and could become useful for prognostic risk stratification and treatment decisions.
- Published
- 2021
42. Activation of autophagy reverses gemcitabine-induced immune inhibition of RAW264.7 macrophages by promoting TNF-α, IL-6 and MHC-II expression
- Author
-
Shanshan Jiang, Li Ye, Yongfang Yuan, Shengnan Li, Kudelaidi Kuerban, Wang Rong, Shu Pan, and Lu Han
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Antimetabolites, Antineoplastic ,Necrosis ,Lipopolysaccharide ,Phagocytosis ,Immunology ,Deoxycytidine ,Proinflammatory cytokine ,Interferon-gamma ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Immune inhibition ,Autophagy ,medicine ,Animals ,Interleukin 6 ,biology ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Cell Cycle ,Histocompatibility Antigens Class II ,Inflammatory cytokines ,Gemcitabine ,Molecular biology ,RAW 264.7 Cells ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Original Article ,Cytokine secretion ,medicine.symptom ,Reactive Oxygen Species ,Immunosuppressive Agents ,RAW264.7 macrophages - Abstract
This research aims to investigate the effect of gemcitabine (GEM) on various activities and functions of macrophages. Phagocytosis, cell autophagy and reactive oxygen species (ROS) were analysed by laser scanning confocal microscope. The cell cycle status and major histocompatibility complex II (MHC-II) expression were examined by flow cytometry. Inflammatory cytokine secretion such as tumour necrosis factor α (TNF-α) and interleukin 6 (IL-6) was detected by Elisa assay. The expression of proteins was analysed by western blot method. The results revealed that GEM-induced immune inhibition of M1-type RAW264.7 macrophages activated by interferon-γ (IFN-γ) and lipopolysaccharide (LPS). We also found that GEM inhibited autophagy, as evidenced by the reduced formation of autophagosome-like vacuoles and autophagosomes. Further study showed that incubation of activated macrophages with the autophagy inhibitor 3-MA induced immune suppression. In contrast, treatment with the autophagy inducer trehalose (Tre) restored phagocytosis, TNF-α and IL-6 secretion, and MHC-II expression in GEM-induced immune-inhibited macrophages. GEM reduced immune effect of M1-type RAW264.7 macrophages via inhibiting TNF-α, IL-6 and MHC-II expression. Furthermore, activation of autophagy by Tre reversed GEM-induced immune inhibition of RAW264.7 macrophages. Supplementary Information The online version contains supplementary material available at 10.1007/s12026-021-09210-7.
- Published
- 2021
43. Efficacy of S‐1 after pemetrexed in patients with non‐small cell lung cancer: A retrospective multi‐institutional analysis
- Author
-
Hiroshi Mukae, Noritaka Honda, Nanae Sugasaki, Hiromi Tomono, Shinnosuke Takemoto, Yasuhiro Umeyama, Hiroyuki Yamaguchi, Sawana Ono, Daiki Ogawara, Katsumi Nakatomi, Hiroaki Senju, Kazumasa Akagi, Yosuke Dotsu, Hirokazu Taniguchi, Hiroshi Gyotoku, Takayuki Suyama, and Minoru Fukuda
- Subjects
Adult ,Male ,Pulmonary and Respiratory Medicine ,Oncology ,Antimetabolites, Antineoplastic ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Antineoplastic Agents ,Pemetrexed ,Disease-Free Survival ,cross‐resistance ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Clinical endpoint ,Humans ,Lung cancer ,RC254-282 ,Aged ,Retrospective Studies ,Tegafur ,Aged, 80 and over ,Chemotherapy ,S‐1 ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Retrospective cohort study ,Original Articles ,General Medicine ,Middle Aged ,medicine.disease ,Confidence interval ,lung cancer ,Drug Combinations ,Oxonic Acid ,Regimen ,Adenocarcinoma ,Original Article ,Female ,business ,medicine.drug - Abstract
Background S‐1 and pemetrexed (PEM) are key treatments for non‐small cell lung cancer (NSCLC). However, the mechanism of anticancer activity of S‐1 and PEM is similar. Cross‐resistance between S‐1 and PEM is of concern. This exploratory study was designed to evaluate the treatment effect of S‐1 following PEM‐containing treatment. Methods This retrospective study included patients with advanced (c‐stage III or IV, UICC seventh edition) or recurrent NSCLC who received S‐1 monotherapy following the failure of previous PEM‐containing chemotherapy at six hospitals in Japan. The primary endpoint of the study was the overall response rate (ORR). The secondary endpoint was the disease control rate (DCR), time to treatment failure (TTF), progression‐free survival (PFS), and overall survival (OS). Results A total of 53 NSCLC patients met the criteria for inclusion in the study. Forty‐six patients had adenocarcinoma (88.7%) and no patients had squamous cell carcinoma. Thirty‐one patients (58.5%) received the standard S‐1 regimen and 18 patients (34.0%) received the modified S‐1 regimen. ORR was 1.9% (95% confidence interval [CI]: 0.00%–10.1%). Median TTF, PFS, and OS were 65, 84, and 385 days, respectively. Conclusions Although there were several limitations in this study, the ORR of S‐1 after PEM in patients with nonsquamous (non‐SQ) NSCLC was low compared to the historical control. One of the options in the future might be to avoid S‐1 treatment in PEM‐treated patients who need tumor shrinkage., Predictive factor of S‐1 and PEM is TS expression.Cross‐resistance between S‐1 and PEM is of concern.This study was designed to evaluate the treatment effect of S‐1 following PEM‐containing treatment.ORR of S‐1 after PEM was low.
- Published
- 2021
44. Visualization of the cancer cell cycle by tissue‐clearing technology using the Fucci reporter system
- Author
-
Kohei Miyazono, Kei Takahashi, Shogo Ehata, Hiroki R. Ueda, Shimpei I. Kubota, Ryo Tanabe, and Yasuyuki Morishita
- Subjects
Antimetabolites, Antineoplastic ,Cancer Research ,Lung Neoplasms ,Genetic Vectors ,Mice, Nude ,Cancer metastasis ,Adenocarcinoma of Lung ,Breast Neoplasms ,Biology ,Transfection ,Metastasis ,Mice ,Cell, Molecular, and Stem Cell Biology ,Genes, Reporter ,In vivo ,Fucci system ,medicine ,Animals ,Humans ,metastasis ,Cell Proliferation ,Mice, Inbred BALB C ,Tissue clearing ,tissue‐clearing technology ,Cell Cycle ,Ubiquitination ,Original Articles ,General Medicine ,Cell cycle ,antitumor drug ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,Luminescent Proteins ,Microscopy, Fluorescence ,Oncology ,A549 Cells ,Luminescent Measurements ,Cancer cell ,Cancer research ,Female ,Original Article ,Fluorouracil ,Imaging technique - Abstract
Tissue‐clearing technology is an emerging imaging technique currently utilized not only in neuroscience research but also in cancer research. In our previous reports, tissue‐clearing methods were used for the detection of metastatic tumors. Here, we showed that the cell cycles of primary and metastatic tumors were visualized by tissue‐clearing methods using a reporter system. First, we established cancer cell lines stably expressing fluorescent ubiquitination‐based cell cycle indicator (Fucci) reporter with widely used cancer cell lines A549 and 4T1. Fluorescence patterns of the Fucci reporter were investigated in various tumor inoculation models in mice. Interestingly, fluorescence patterns of the Fucci reporter of tumor colonies were different between various organs, and even among colonies in the same organs. The effects of antitumor drugs were also evaluated using these Fucci reporter cells. Of the three antitumor drugs studied, 5‐fluorouracil treatment on 4T1‐Fucci cells resulted in characteristic fluorescent patterns by the induction of G2/M arrest both in vitro and in vivo. Thus, the combination of a tissue‐clearing method with the Fucci reporter is useful for analyzing the mechanisms of cancer metastasis and drug resistance., Monitoring the Fucci patterns in vivo with 4T1‐Fucci cells. A, B, 3D and 2D images of 4T1 cells inoculated orthotopically into the mammary fat‐pad. C, 3D and 2D images of 4T1‐Fucci cells in experimental lung metastasis.
- Published
- 2021
45. Age as a modifier of the effects of chemoradiotherapy with infusional 5-fluorouracil after D2 dissection in gastric cancer
- Author
-
Chao-Hsun Chen, Sung-Wei Lee, Chien-Liang Lin, Hung-Chang Wu, Wen-Tsung Huang, Yan-Xun Chen, Chao-Jung Tsao, Shang-Wen Chen, How Ran Guo, Wen-Li Lin, Cheng-Yao Lin, and Shang-Hung Chen
- Subjects
Adult ,Male ,Antimetabolites, Antineoplastic ,Aging ,medicine.medical_specialty ,medicine.medical_treatment ,Taiwan ,D2 dissection ,elderly ,Gastroenterology ,Disease-Free Survival ,chemoradiotherapy ,Stomach Neoplasms ,Internal medicine ,medicine ,Humans ,Infusions, Intravenous ,Lymph node ,Aged ,Aged, 80 and over ,Chemotherapy ,business.industry ,gastric cancer ,Hazard ratio ,Cancer ,Chemoradiotherapy, Adjuvant ,Cell Biology ,Middle Aged ,Prognosis ,medicine.disease ,Survival Analysis ,Radiation therapy ,Treatment Outcome ,medicine.anatomical_structure ,Fluorouracil ,Lymph Node Excision ,Female ,infusional 5-fluorouracil ,business ,Adjuvant ,Chemoradiotherapy ,Research Paper ,medicine.drug - Abstract
Adjuvant concurrent chemoradiotherapy (CCRT) is the standard care for patients with resected advanced gastric cancer, but its survival benefits remain undetermined in patients undergoing D2 lymph node dissection (D2 dissection). We evaluated safety and efficacy of adjuvant CCRT with 5-fluorouracil (5-FU) versus chemotherapy alone in 110 gastric cancer patients with D2 dissection treated in Taiwan between January 2009 and January 2013. All the 71 patients receiving adjuvant CCRT were treated with daily infusional 5-FU and radiotherapy. Adjuvant CCRT was associated with higher risks of major hematologic (56.3% vs. 23.8%, p = 0.002) and gastrointestinal (46.9% vs. 14.3%, p = 0.027) toxicities and death (12.5% vs. 0.0%, p = 0.041) in patients above 70 years old, but this was not the case in those ≤70 years of age. Univariate Cox proportional regressions identified adjuvant CCRT as a factor for better overall survival (OS) (hazard ratio [HR]=0.52; 95% confidence interval [CI]: 0.27–0.99) and disease-free survival (DFS) (HR=0.46, 95% CI: 0.24–0.88), but it was not a significant factor for OS or DFS after adjusting for other factors in the multivariate analysis. However, in stratified analyses by age, we found adjuvant CCRT was an independent prognostic factor for better OS (HR=0.07; 95% CI: 0.01–0.38) in patients ≤70 years old, but not in those above 70 years of age. Therefore, it was concluded that age may to be a modifier of the effects of adjuvant CCRT.
- Published
- 2021
46. Monitoring Of High-Dose Methotrexate (Mtx)-Related Toxicity and Mtx Levels in Children with Acute Lymphoblastic Leukemia: A Pilot-Study in Indonesia
- Author
-
Zulfan Zazuli, Muhammad Hasan Bashari, Eddy Supriyadi, Susi Susanah, Nur Suryawan, Harry Raspati, Gertjan J.L. Kaspers, Ponpon Idjradinata, Lelani Reniarti, Lulu Eva Rakhmilla, Nur Melani Sari, Pediatrics, CCA - Cancer Treatment and quality of life, and CCA - Cancer biology and immunology
- Subjects
Male ,musculoskeletal diseases ,Antimetabolites, Antineoplastic ,medicine.medical_specialty ,Drug-Related Side Effects and Adverse Reactions ,Population ,Pilot Projects ,Acute lymphoblastic leukemia ,Neutropenia ,Loading dose ,Gastroenterology ,Nephrotoxicity ,Internal medicine ,high dose methotrexate ,Mucositis ,Humans ,Medicine ,Prospective Studies ,Child ,Infusions, Intravenous ,education ,education.field_of_study ,2013 ALL Indonesian protocol ,business.industry ,toxicity ,General Medicine ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Methotrexate ,Indonesia ,Toxicity ,Female ,business ,Febrile neutropenia ,Research Article ,medicine.drug - Abstract
The administration of high-dose methotrexate (HD-MTX) requires an accurate monitoring of blood MTX levels to determine the regimen of leucovorin rescue and urine alkalinization to prevent toxicity. However, it is technically and logistically challenging to screen patients routinely in limited-resource settings. This study aimed to evaluate blood MTX levels at 24- and 48-hours from start of infusion in relation to clinical toxicity in childhood ALL. Methods: A prospective cohort study was conducted on 32 consecutive children with acute lymphoblastic leukemia (ALL) who had received at least one cycle of 1 g/m2 HD-MTX intravenous infusion as a part of consolidation treatment based on the 2013 Indonesian ALL Protocol. In total, 68 cycles were evaluated. Serum MTX concentrations were measured using enzyme immunoassay. MTX toxicity was categorized using common toxicity criteria (CTCAE) 3.0 version. The association between MTX level and clinical toxicity was assessed by non-parametric analysis. Results: The 24-hours MTX level was median 29.8 ng/mL (0.065 µmol/L) (IQR 8.1–390.6) with a modest decrease in 48-hours MTX serum level in all cycles (median 28.3 ng/mL and 0.062 µmol/L; IQR 0.35–28.7; p
- Published
- 2021
47. 5-aza-2′-deoxycitidine inhibits cell proliferation, extracellular matrix formation and Wnt/β-catenin pathway in human uterine leiomyomas
- Author
-
Julia Escrig, María Cristina Carbajo-García, Irene Cervelló, Ana Corachán, Hortensia Ferrero, Marina Segura-Benitez, Javier Monleón, Antonio Pellicer, and Amparo Faus
- Subjects
Adult ,0301 basic medicine ,Antimetabolites, Antineoplastic ,QH471-489 ,Cell Survival ,Uterine leiomyoma ,Decitabine ,MMP7 ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,5-aza-2′-deoxycitidine ,Humans ,Wnt/β-catenin pathway ,Prospective Studies ,DNA Methylation Inhibition ,Wnt Signaling Pathway ,Cell proliferation ,Dose-Response Relationship, Drug ,Leiomyoma ,biology ,Chemistry ,Research ,Reproduction ,Wnt signaling pathway ,Myometrium ,Obstetrics and Gynecology ,Gynecology and obstetrics ,DNA Methylation ,Middle Aged ,Extracellular Matrix ,Proliferating cell nuclear antigen ,030104 developmental biology ,Reproductive Medicine ,030220 oncology & carcinogenesis ,Catenin ,Uterine Neoplasms ,DNA methylation ,biology.protein ,Cancer research ,RG1-991 ,Female ,Epigenetics ,Developmental Biology - Abstract
Background Uterine leiomyoma is a benign tumor with unclear pathogenesis and inaccurate treatment. This tumor exhibits altered DNA methylation related to disease progression. DNMT inhibitors as 5-aza-2′-deoxycytidine (5-aza-CdR), have been suggested to treat tumors in which DNA methylation is altered. We aimed to evaluate whether DNA methylation reversion with 5-aza-CdR reduces cell proliferation and extracellular matrix (ECM) formation in uterine leiomyoma cells to provide a potential treatment option. Methods Prospective study using uterine leiomyoma and adjacent myometrium tissues and human uterine leiomyoma primary (HULP) cells (n = 16). In tissues, gene expression was analyzed by qRT-PCR and DNMT activity by ELISA. Effects of 5-aza-CdR treatment on HULP cells were assessed by CellTiter, western blot, and qRT-PCR. Results DNMT1 gene expression was higher in uterine leiomyoma vs myometrium. Similarly, DNMT activity was greater in uterine leiomyoma and HULP cells (6.5 vs 3.8 OD/h/mg; 211.3 vs 63.7 OD/h/mg, respectively). After 5-aza-CdR treatment on HULP cells, cell viability was reduced, significantly so at 10 μM (85.3%). Treatment with 10 μM 5-aza-CdR on HULP cells significantly decreased expression of proliferation marker PCNA (FC = 0.695) and of ECM proteins (COLLAGEN I FC = 0.654; PAI-1, FC = 0.654; FIBRONECTIN FC = 0.733). 5-aza-CdR treatment also decreased expression of Wnt/β-catenin pathway final targets, including WISP1 protein expression (10 μM, FC = 0.699), c-MYC gene expression (2 μM, FC = 0.745 and 10 μM, FC = 0.728), and MMP7 gene expression (5 μM, FC = 0.520 and 10 μM, FC = 0.577). Conclusions 5-aza-CdR treatment inhibits cell proliferation, ECM formation, and Wnt/β-catenin signaling pathway targets in HULP cells, suggesting that DNA methylation inhibition is a viable therapeutic target in uterine leiomyoma.
- Published
- 2021
48. Exosomal annexin A6 induces gemcitabine resistance by inhibiting ubiquitination and degradation of EGFR in triple-negative breast cancer
- Author
-
Biyun Wang, Xiaojia Liu, Ting Li, Jun Cao, Leiping Wang, Zhonghua Tao, Yiqun Du, Xichun Hu, Yihui Zhu, and Jian Zhang
- Subjects
0301 basic medicine ,Cancer Research ,Antimetabolites, Antineoplastic ,Immunology ,Apoptosis ,Triple Negative Breast Neoplasms ,Lapatinib ,Predictive markers ,Exosomes ,Deoxycytidine ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Breast cancer ,Annexin ,Cell Line, Tumor ,medicine ,Humans ,Viability assay ,Annexin A6 ,Triple-negative breast cancer ,Cell Proliferation ,Tumor microenvironment ,QH573-671 ,Chemistry ,Ubiquitination ,Cell Biology ,Gemcitabine ,Microvesicles ,ErbB Receptors ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer cell ,Proteolysis ,Cancer research ,Cytology ,medicine.drug - Abstract
Exosomes are carriers of intercellular information that regulate the tumor microenvironment, and they have an essential role in drug resistance through various mechanisms such as transporting RNA molecules and proteins. Nevertheless, their effects on gemcitabine resistance in triple-negative breast cancer (TNBC) are unclear. In the present study, we examined the effects of exosomes on TNBC cell viability, colony formation, apoptosis, and annexin A6 (ANXA6)/EGFR expression. We addressed their roles in gemcitabine resistance and the underlying mechanism. Our results revealed that exosomes derived from resistant cancer cells improved cell viability and colony formation and inhibited apoptosis in sensitive cancer cells. The underlying mechanism included the transfer of exosomal ANXA6 from resistant cancer cells to sensitive cancer cells. Isobaric peptide labeling–liquid chromatography–tandem mass spectrometry and western blotting revealed that ANXA6 was upregulated in resistant cancer cells and their derived exosomes. Sensitive cancer cells exhibited resistance with increased viability and colony formation and decreased apoptosis when ANXA6 was stably overexpressed. On the contrary, knockdown ANXA6 restored the sensitivity of cells to gemcitabine. Co-immunoprecipitation expression and GST pulldown assay demonstrated that exosomal ANXA6 and EGFR could interact with each other and exosomal ANXA6 was associated with the suppression of EGFR ubiquitination and downregulation. While adding lapatinib reversed gemcitabine resistance induced by exosomal ANXA6. Moreover, ANXA6 and EGFR protein expression was correlated in TNBC tissues, and exosomal ANXA6 levels at baseline were lower in patients with highly sensitive TNBC than those with resistant TNBC when treated with first-line gemcitabine-based chemotherapy. In conclusion, resistant cancer cell-derived exosomes induced gemcitabine resistance via exosomal ANXA6, which was associated with the inhibition of EGFR ubiquitination and degradation. Exosomal ANXA6 levels in the serum of patients with TNBC might be predictive of the response to gemcitabine-based chemotherapy.
- Published
- 2021
49. mTORC2 regulates ribonucleotide reductase to promote DNA replication and gemcitabine resistance in non-small cell lung cancer
- Author
-
Caiyong Lai, Guo Chen, Yan Chen, Naite Xi, Ling Tian, Mingrong Cao, Yongchang Zhang, Qi Feng, Congcong Chen, Shengbin Lin, Yanguan Guo, Li Yu, Jiaxin Tian, Jinye Mi, Jun Fan, and Fan Zhang
- Subjects
0301 basic medicine ,DNA Replication ,Cancer Research ,Antimetabolites, Antineoplastic ,Ribonucleoside Diphosphate Reductase ,DNA damage ,Cell ,Mechanistic Target of Rapamycin Complex 2 ,Deoxycytidine ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Ribonucleotide Reductases ,medicine ,EGFR, Epidermal growth factor receptor ,Humans ,Phosphorylation ,NSCLC, Non-small cell lung cancer ,dNTP, deoxyribonucleotide triphosphates ,Ribonucleotide reductase ,mTORC2 ,RC254-282 ,Original Research ,Chemistry ,DNA replication ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,DNA Replication Fork ,DSB, Double-strand break ,Gemcitabine ,DNA replication stress ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Cancer cell ,Cancer research ,RNR, Ribonucleotide reductase ,medicine.drug ,DNA Damage ,Protein Binding ,Signal Transduction - Abstract
Ribonucleotide reductase (RNR) is the key enzyme that catalyzes the production of deoxyribonucleotides (dNTPs) for DNA replication and it is also essential for cancer cell proliferation. As the RNR inhibitor, Gemcitabine is widely used in cancer therapies, however, resistance limits its therapeutic efficacy and curative potential. Here, we identified that mTORC2 is a main driver of gemcitabine resistance in non-small cell lung cancers (NSCLC). Pharmacological or genetic inhibition of mTORC2 greatly enhanced gemcitabine induced cytotoxicity and DNA damage. Mechanistically, mTORC2 directly interacted and phosphorylated RNR large subunit RRM1 at Ser 631. Ser631 phosphorylation of RRM1 enhanced its interaction with small subunit RRM2 to maintain sufficient RNR enzymatic activity for efficient DNA replication. Targeting mTORC2 retarded DNA replication fork progression and improved therapeutic efficacy of gemcitabine in NSCLC xenograft model in vivo. Thus, these results identified a mechanism through mTORC2 regulating RNR activity and DNA replication, conferring gemcitabine resistance to cancer cells.
- Published
- 2021
50. Low‐dose decitabine plus venetoclax is safe and effective as post‐transplant maintenance therapy for high‐risk acute myeloid leukemia and myelodysplastic syndrome
- Author
-
Xin Li, Ting Yuan, Rui Sun, Xia Xiong, Yunxiong Wei, Hairong Lyu, Xin Jin, Ming-Feng Zhao, Wenyi Lu, Xiaoyuan He, and Tongtong Sun
- Subjects
Oncology ,Male ,Cancer Research ,Transplantation Conditioning ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,chemistry.chemical_compound ,Maintenance therapy ,Recurrence ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Cumulative incidence ,Prospective Studies ,Sulfonamides ,Hematopoietic Stem Cell Transplantation ,General Medicine ,Middle Aged ,Progression-Free Survival ,Leukemia, Myeloid, Acute ,surgical procedures, operative ,Tolerability ,Original Article ,Female ,medicine.drug ,Adult ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,Decitabine ,Neutropenia ,Venetoclax ,Young Adult ,Clinical Research ,Internal medicine ,Humans ,Transplantation, Homologous ,Aged ,business.industry ,Original Articles ,allo‐HSCT ,medicine.disease ,Bridged Bicyclo Compounds, Heterocyclic ,maintence therapy ,chemistry ,Hypomethylating agent ,Myelodysplastic Syndromes ,business ,Follow-Up Studies - Abstract
Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) are usually associated with poor outcomes, especially in high‐risk AML/MDS. Allogeneic hematopoietic stem cell transplantation (allo‐HSCT) is the only curative option for patients suffering from high‐risk AML/MDS. However, many patients relapse after allo‐HSCT. Novel therapy to prevent relapse is urgently needed. Both the BCL‐2 inhibitor venetoclax (VEN) and the hypomethylating agent decitabine (DEC) possess significant antitumor activity effects against AML/MDS. Administration of DEC has been shown to ameliorate graft‐versus‐host disease (GVHD) and boost the graft‐versus‐leukemia (GVL) effect post‐transplantation. We therefore conducted a prospective study (ChiCTR1900025374) to examine the tolerability and efficacy of a maintenance therapy of low‐dose decitabine (LDEC) plus VEN to prevent relapse after allo‐HSCT for high‐risk AML/MDS patients. Twenty patients with high‐risk AML (n = 17) or high‐risk MDS (n = 3) post‐transplantation were recruited. Approximately day 100 post‐transplantation, all patients received LDEC (15 mg/m2 for 3 d) followed by VEN (200 mg) on d 1‐21. The cycle interval was 2 mo, and there was 10 cycles. The primary end points of this study were rates of overall survival (OS) and event‐free survival (EFS). The secondary endpoints included adverse events (AEs), cumulative incidence of relapse (CIR), nonrelapse mortality (NRM), incidences of acute GVHD (aGVHD) and chronic GVHD (cGVHD), and incidences of viral infection after allo‐HSCT. Survival outcomes were assessed using Kaplan‐Meier analysis. The median follow‐up was 598 (149‐1072) d. Two patients relapsed, 1 died, and 1 is still alive after the second transplant. The 2‐y OS and EFS rates were 85.2% and 84.7%, respectively. The median 2‐y EFS time was 525 (149‐1072) d, and 17 patients still had EFS and were alive at the time of this writing. The most common AEs were neutropenia, anemia, thrombocytopenia, neutropenic fever, and fatigue. Grade 2 or 3 AEs were observed in 35% (7/20) and 20% (4/20) of the patients, respectively. No grade >3 AEs were observed. aGVHD (any grade) and cGVHD (limited or extensive) occurred in 55% and 20% of patients, respectively. We conclude that LDEC + VEN can be administered safely after allo‐HSCT with no evidence of an increased incidence of GVHD, and this combination decreases the relapse rate in high‐risk AML/MDS patients. This novel maintenance therapy may be a promising way to prevent relapse in high‐risk AML/MDS patients., The results of the current study suggest that maintenance treatment with LDEC combined with VEN introduced nearly 3 mo after allo‐HSCT is efficacious, with an acceptable toxicity profile and impressive long‐term disease control.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.