8,221 results on '"TEMOZOLOMIDE"'
Search Results
2. Investigators at AntiCancer Inc. Discuss Findings in Gliomas (Extensive Shrinkage and Long-term Stable Disease In a Teenage Female Patient With High-grade Glioma Treated With Temozolomide and Radiation In Combination With Oral Recombinant...).
- Subjects
TEMOZOLOMIDE ,GLIOMAS ,SULFUR amino acids ,ESSENTIAL amino acids ,WOMEN patients ,BRAIN tumors - Abstract
A recent study conducted by investigators at AntiCancer Inc. in San Diego, California, has found that methionine restriction, combined with radiation and temozolomide (TMZ) chemotherapy, was highly effective in treating a teenage female patient with high-grade glioma. Gliomas are malignant primary brain tumors that are difficult to treat. The patient's tumor size shrunk by at least 60% after the start of methionine restriction, and her condition remained stable for 19 months without severe adverse effects. This research provides promising results for the treatment of gliomas using this combination therapy. [Extracted from the article]
- Published
- 2024
3. Radiation-induced intracranial rhabdomyosarcoma- A rare complication: Report of a case with literature review
- Author
-
Apinderpreet Singh, Renu Madan, Bishan Dass Radotra, Chirag Komal Ahuja, Debajyoti Chatterjee, and Geethanjali Gude
- Subjects
medicine.medical_specialty ,Chemotherapy ,Temozolomide ,Gliosarcoma ,business.industry ,medicine.medical_treatment ,General Medicine ,medicine.disease ,Lesion ,Radiation therapy ,medicine ,Embryonal rhabdomyosarcoma ,Radiology ,Sarcoma ,medicine.symptom ,Complication ,business ,medicine.drug - Abstract
Radiation-induced sarcoma (RIS) of the central nervous system is an uncommon late complication of radiation therapy. We report a case of a 47-year-old male patient who underwent surgery followed by irradiation and chemotherapy with temozolomide for a frontal lobe gliosarcoma and presented 43 months later with a recurrent tumor in the same location with interval growth in the size of the lesion. Histology from surgical resection of the recurrent tumor revealed embryonal rhabdomyosarcoma (RMS). Adjacent brain parenchyma showed radiation-induced changes. There was no evidence of gliosarcoma at recurrence. In addition to the rarity of sarcomas arising following irradiation for glial tumors, this case represents one of the first reports of an intracerebral RMS arising in this setting.
- Published
- 2023
4. Stereotactic radiosurgery for glioblastoma considering tumor genetic profiles: an international multicenter study
- Author
-
Narendra Kumar, Erik P. Sulman, David E. Arsanious, Angel Mota, Herwin Speckter, Dev N Patel, Joshua S Weir, Kenneth Bernstein, Jason P. Sheehan, Cheng-Chia Lee, Huai-Che Yang, David Mathieu, Stylianos Pikis, Basem Dahshan, Douglas Kondziolka, Violaine Delabar, Ronald E Warnick, Adomas Bunevicius, Manjul Tripathi, and Christopher P. Cifarelli
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,Temozolomide ,Methyltransferase ,business.industry ,medicine.medical_treatment ,General Medicine ,medicine.disease ,Radiosurgery ,Isocitrate dehydrogenase ,Multicenter study ,Internal medicine ,medicine ,business ,Median survival ,medicine.drug ,Glioblastoma - Abstract
OBJECTIVE Molecular profiles, such as isocitrate dehydrogenase (IDH) mutation and O6-methylguanine-DNA methyltransferase (MGMT) methylation status, have important prognostic roles for glioblastoma patients. The authors studied the efficacy and safety of stereotactic radiosurgery (SRS) for glioblastoma patients with consideration of molecular tumor profiles. METHODS For this retrospective observational multiinstitutional study, the authors pooled consecutive patients who were treated using SRS for glioblastoma at eight institutions participating in the International Radiosurgery Research Foundation. They evaluated predictors of overall and progression-free survival with consideration of IDH mutation and MGMT methylation status. RESULTS Ninety-six patients (median age 56 years) underwent SRS (median dose 15 Gy and median treatment volume 5.53 cm3) at 147 tumor sites (range 1 to 7). The majority of patients underwent prior fractionated radiation therapy (92%) and temozolomide chemotherapy (98%). Most patients were treated at recurrence (85%), and boost SRS was used for 12% of patients. The majority of patients harbored IDH wild-type (82%) and MGMT-methylated (62%) tumors. Molecular data were unavailable for 33 patients. Median survival durations after SRS were similar between patients harboring IDH wild-type tumors and those with IDH mutant tumors (9.0 months vs 11 months, respectively), as well as between those with MGMT-methylated tumors and those with MGMT-unmethylated tumors (9.8 vs. 9.0 months, respectively). Prescription dose > 15 Gy (OR 0.367, 95% CI 0.190–0.709, p = 0.003) and treatment volume > 5 cm3 (OR 1.036, 95% CI 1.007–1.065, p = 0.014) predicted overall survival after controlling for age and IDH status. Treatment volume > 5 cm3 (OR 2.215, 95% CI 1.159–4.234, p = 0.02) and absence of gross-total resection (OR 0.403, 95% CI 0.208–0.781, p = 0.007) were associated with inferior local control of SRS-treated lesions in multivariate models. Nine patients experienced adverse radiation events after SRS, and 7 patients developed radiation necrosis at 59 to 395 days after SRS. CONCLUSIONS Post-SRS survival was similar as a function of IDH mutation and MGMT promoter methylation status, suggesting that molecular profiles of glioblastoma should be considered when selecting candidates for SRS. SRS prescription dose > 15 Gy and treatment volume ≤ 5 cm3 were associated with longer survival, independent of age and IDH status. Prior gross-total resection and smaller treatment volume were associated with superior local control.
- Published
- 2022
5. Influence of Concurrent and Adjuvant Temozolomide on Health-Related Quality of Life of Patients with Grade III Gliomas: A Secondary Analysis of a Randomized Clinical Trial (KNOG-1101 Study)
- Author
-
Grace S. Ahn, Heon Yoo, Jinhee Kim, Chul-Kee Park, Tae Min Kim, Se-Hyuk Kim, Jeong Hoon Kim, Gheeyoung Choe, Yong-Kil Hong, Kihwan Hwang, Do-Hyun Nam, Yu Jung Kim, Tae-Young Jung, Jong Hee Chang, Chae-Yong Kim, Jungnam Joo, Byung Se Choi, Dong-Eun Lee, Eun Young Kim, and Seok Gu Kang
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Nausea ,medicine.medical_treatment ,law.invention ,Quality of life ,Randomized controlled trial ,law ,Internal medicine ,Temozolomide ,medicine ,WHO Grade III Glioma ,Humans ,Lymphoma, Follicular ,Chemotherapy ,Brain Neoplasms ,business.industry ,Chemoradiotherapy ,Glioma ,humanities ,Radiation therapy ,Quality of Life ,Vomiting ,medicine.symptom ,business ,medicine.drug - Abstract
PurposeThe KNOG-1101 study showed improved 2-year PFS with temozolomide during and after radiotherapy compared to radiotherapy alone for patients with anaplastic gliomas. This trial investigates the effect of concurrent and adjuvant temozolomide on health-related quality of life (HRQoL).Materials and MethodsIn this randomized, open-label, phase II trial, 90 patients with World Health Organization grade III glioma were enrolled across multiple centers in South Korea between March 2012 to February 2015 and followed up through 2017. The European Organization for Research and Treatment of Cancer Quality of Life Questionnaire 30 (EORTC QLQ-C30) and 20-item EORTC QLQ-Brain Neoplasm (QLQ-BN20) were used to compare HRQoL between patients assigned to concurrent chemoradiotherapy with temozolomide followed by 6 cycles of adjuvant temozolomide (arm A) and radiotherapy (RT) alone (arm B).ResultsOf the 90 patients in the study, 84 patients (93.3%) completed the baseline HRQoL questionnaire. Emotional functioning, fatigue, nausea and vomiting, dyspnea, constipation, appetite loss, diarrhea, seizures, itchy skin, drowsiness, hair loss, and bladder control were not affected by the addition of temozolomide. All other items did not differ significantly between arm A and arm B throughout treatment. Global health status particularly stayed consistent at the end of adjuvant temozolomide (p=0.47) and at the end of RT (p=0.33).ConclusionThe addition of concurrent and adjuvant temozolomide did not show negative influence on HRQoL with improvement of progression-free survival for patients with anaplastic gliomas. The absence of systematic and clinically relevant changes in HRQoL suggests that an overall long-term net clinical benefit exists for concurrent and adjuvant temozolomide.
- Published
- 2022
6. Wnt/β-catenin Antagonists: Exploring New Avenues to Trigger Old Drugs in Alleviating Glioblastoma Multiforme
- Author
-
Parthiban Marimuthu, Thirugnanasambandham Sivasubramanian Anitha, Mugilarasi Purushothaman, Muralidharan A Ramachandran, Daisy S. Precilla, and Shreyas S Kuduvalli
- Subjects
Temozolomide ,Combination therapy ,business.industry ,Wnt signaling pathway ,Context (language use) ,General Medicine ,Drug repositioning ,Cell Line, Tumor ,Catenin ,medicine ,Cancer research ,Humans ,Signal transduction ,Glioblastoma ,business ,Receptor ,Wnt Signaling Pathway ,beta Catenin ,Cell Proliferation ,medicine.drug - Abstract
Background: Glioblastoma Multiforme (GBM) is one of the most heterogeneous primary brain tumors with high mortality. In spite of the current therapeutic approaches, the survival rate remains poor, with death occurring within 12 to 15 months after the preliminary diagnosis. This warrants the need for an effective treatment modality. The Wnt/β-catenin pathway is presumably the most noteworthy pathway upregulated in almost 80% of GBM cases, contributing to tumor initiation, progression, and survival. Therefore, therapeutic strategies targeting key components of the Wnt/β-catenin cascade using established genotoxic agents like temozolomide and pharmacological inhibitors would be an effective approach to modulate the Wnt/β-catenin pathway. Recently, drug repurposing by means of effective combination therapy has gained importance in various solid tumors, including GBM, by targeting two or more proteins in a single pathway, thereby possessing the ability to overcome the hurdle implicated by chemoresistance in GBM. Objective: In this context, by employing computational tools, an attempt has been made to find out the novel combinations against the Wnt/β-catenin signalling pathway. Methods: We have explored the binding interactions of three conventional drugs - namely temozolomide, metformin and chloroquine - along with three natural compounds, viz. epigallocatechin gallate, naringenin and phloroglucinol, on the major receptors of Wnt/β-catenin signalling. Results: It was noted that all the experimental compounds showed profound interaction with two major receptors of the Wnt/β-catenin pathway. Conclusion: To the best of our knowledge, this study is the first of its kind to characterize the combined interactions of the aforementioned drugs with the Wnt/β-catenin signalling in silico, and this will putatively open up new avenues for combination therapies in GBM treatment.
- Published
- 2022
7. Sequential Capecitabine/Temozolomide and Sunitinib Treatment in Patients With Metastatic Well-Differentiated Grade 1/Grade 2 Pancreatic Neuroendocrine Tumors
- Author
-
Wenquan Wang, Hao Li, Wu-Hu Zhang, He-Li Gao, Long-Yun Ye, Quanxing Ni, Hua-Xiang Xu, Jia Dong, and Liang Liu
- Subjects
Oncology ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Neuroendocrine tumors ,urologic and male genital diseases ,Capecitabine ,Endocrinology ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Sunitinib ,Temozolomide ,Humans ,Medicine ,Progression-free survival ,Chemoembolization, Therapeutic ,Prospective cohort study ,Transcatheter arterial chemoembolization ,Retrospective Studies ,business.industry ,General Medicine ,medicine.disease ,female genital diseases and pregnancy complications ,Pancreatic Neoplasms ,Neuroendocrine Tumors ,Tolerability ,business ,medicine.drug - Abstract
Objective The role of alternate sequential administration of sunitinib and capecitabine/temozolomide (CAPTEM) in metastatic pancreatic neuroendocrine tumors (PanNETs) remains unexplored. We thus aimed to analyze the efficacy and tolerability of this strategy in advanced grade 1/grade 2 PanNETs. Methods In total, data of 43 patients with metastatic PanNET were collected from a real-world database of a cancer center. Twenty-four patients were treated with sunitinib followed by CAPTEM (group 1), and 19 patients were treated with CAPTEM followed by sunitinib (group 2). Results Twenty-three patients were treated with first-line sunitinib or CAPTEM, and 20 patients were pretreated with somatostatin analog (SSA) or SSA in combination with transcatheter arterial chemoembolization. The objective response rate with first-line treatment was similar in both groups, whereas that with second-line treatment was higher in group 1 than in group 2, albeit with no significant differences (21.1% vs 5.3%, respectively; P = .205). Median progression-free survival (mPFS) for first-line and second-line treatments did not differ between the 2 groups (11 and 12 months vs 12 and 8 months, respectively). Following subgroup analyses, treatment with first-line sunitinib and sunitinib after pretreated SSA had a longer mPFS than that with second-line sunitinib after CAPTEM (11 months vs 8 months, respectively; P = .046), whereas treatment with first-line CAPTEM and CAPTEM after pretreated SSA had an mPFS similar to that of second-line CAPTEM after sunitinib treatment. CAPTEM and sunitinib had similar tolerability. Conclusion Alternating sunitinib and CAPTEM were well tolerated and associated with similar mPFS in grade 1/grade 2 PanNETs. However, larger prospective studies are required to investigate the efficacy of alternate sequential therapies for metastatic PanNET.
- Published
- 2022
8. Sporadic and Lynch syndrome-associated mismatch repair-deficient brain tumors
- Author
-
Hongseok Yun, Seung Hong Choi, Ka Young Lim, Seung-Ki Kim, Yumi Shim, Tae Min Kim, Jin-Woo Park, Hyunhee Kim, Jeongwan Kang, Sung Hye Park, Chul-Kee Park, Kwanghoon Lee, and Jae Kyung Won
- Subjects
Adult ,Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Gliosarcoma ,Adolescent ,Brain tumor ,Gene mutation ,DNA Mismatch Repair ,Article ,Pathology and Forensic Medicine ,Glioma ,Biomarkers, Tumor ,Temozolomide ,Cancer genomics ,medicine ,Humans ,Child ,Antineoplastic Agents, Alkylating ,neoplasms ,Molecular Biology ,Oncogenesis ,Aged ,Brain Neoplasms ,business.industry ,Brain ,High-Throughput Nucleotide Sequencing ,Microsatellite instability ,Astrocytoma ,Cell Biology ,Middle Aged ,medicine.disease ,Colorectal Neoplasms, Hereditary Nonpolyposis ,Magnetic Resonance Imaging ,digestive system diseases ,Lynch syndrome ,Pedigree ,nervous system diseases ,Mutation ,Cancer research ,Female ,business ,medicine.drug - Abstract
Mismatch repair-deficient (MMRD) brain tumors are rare among primary brain tumors and can be induced by germline or sporadic mutations. Here, we report 13 MMRD-associated (9 sporadic and 4 Lynch syndrome) primary brain tumors to determine clinicopathological and molecular characteristics and biological behavior. Our 13 MMRD brain tumors included glioblastoma (GBM) IDH-wildtype (n = 9) including 1 gliosarcoma, astrocytoma IDH-mutant WHO grade 4 (n = 2), diffuse midline glioma (DMG) H3 K27M-mutant (n = 1), and pleomorphic xanthoastrocytoma (PXA) (n = 1). Next-generation sequencing using a brain tumor-targeted gene panel, microsatellite instability (MSI) testing, Sanger sequencing for germline MMR gene mutation, immunohistochemistry of MMR proteins, and clinicopathological and survival analysis were performed. There were many accompanying mutations, suggesting a high tumor mutational burden (TMB) in 77%, but TMB was absent in one case of GBM, IDH-wildtype, DMG, and PXA, respectively. MSH2, MLH1, MSH6, and PMS2 mutations were found in 31%, 31%, 31% and 7% of patients, respectively. MSI-high and MSI-low were found in 50% and 8% of these gliomas, respectively and 34% was MSI-stable. All Lynch syndrome-associated GBMs had MSI-high. In addition, 77% (10/13) had histopathologically multinucleated giant cells. The progression-free survival tended to be poorer than the patients with no MMRD gliomas, but the number and follow-up duration of our patients were insufficient to get statistical significance. In the present study, we found that the most common MMRD primary brain tumor was GBM IDH-wildtype. The genetic profile of MMRD GBM was different from that of conventional GBM. MMRD gliomas with TMB and MSI-H may be sensitive to immunotherapy but resistant to temozolomide. Our findings can help develop better treatment options., The authors examined 13 mismatch repair-deficient (MMRD)-associated primary brain tumors to determine molecular characteristics and biological behavior. MMRD occurred after chemotherapy and radiotherapy in two cases. The genetic profile of MMRD glioblastoma was different from that of conventional glioblastoma. Half of the MMRD-gliomas and all Lynch syndrome-associated GBMs were high in microsatellite instability. These tumors had high mutational burdens and tended to have shorter progression-free survival than glioma without MMRD.
- Published
- 2022
9. Establishment of Patient-Derived Succinate Dehydrogenase–Deficient Gastrointestinal Stromal Tumor Models for Predicting Therapeutic Response
- Author
-
Sunil J. Advani, Hyunho Yoon, Rowan Ustoy, Mayra Yebra, Vipul R. Sheth, Sudeep Banerjee, Michael Heinrich, Christian M. Metallo, Joseph A. Aguilera, Sangkyu Noh, Scott M. Lippman, Razelle Kurzrock, Chih-Min Tang, Olivier Harismendy, Paul T. Fanta, Adam M. Burgoyne, Avi Kumar, Thekla Cordes, Shruti Bhargava, Sam Li, Jason K. Sicklick, and Christopher L. Corless
- Subjects
Cancer Research ,Receptor, Platelet-Derived Growth Factor alpha ,Adolescent ,Gastrointestinal Stromal Tumors ,Oncology and Carcinogenesis ,PDGFRA ,Article ,Young Adult ,03 medical and health sciences ,Rare Diseases ,0302 clinical medicine ,Clinical Research ,Humans ,Medicine ,Oncology & Carcinogenesis ,Stromal tumor ,neoplasms ,Gastrointestinal Neoplasms ,Cancer ,030304 developmental biology ,0303 health sciences ,Gastrointestinal tract ,Temozolomide ,GiST ,business.industry ,Platelet-Derived Growth Factor alpha ,medicine.disease ,digestive system diseases ,3. Good health ,Succinate Dehydrogenase ,Clinical trial ,Proto-Oncogene Proteins c-kit ,Orphan Drug ,Good Health and Well Being ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Sarcoma ,Digestive Diseases ,business ,Tyrosine kinase ,Receptor ,medicine.drug - Abstract
Purpose: Gastrointestinal stromal tumor (GIST) is the most common sarcoma of the gastrointestinal tract, with mutant succinate dehydrogenase (SDH) subunits (A–D) comprising less than 7.5% (i.e., 150–200/year) of new cases annually in the United States. Contrary to GISTs harboring KIT or PDGFRA mutations, SDH-mutant GISTs affect adolescents/young adults, often metastasize, and are frequently resistant to tyrosine kinase inhibitors (TKI). Lack of human models for any SDH-mutant tumors, including GIST, has limited molecular characterization and drug discovery. Experimental Design: We describe methods for establishing novel patient-derived SDH-mutant (mSDH) GIST models and interrogated the efficacy of temozolomide on these tumor models in vitro and in clinical trials of patients with mSDH GIST. Results: Molecular and metabolic characterization of our patient-derived mSDH GIST models revealed that these models recapitulate the transcriptional and metabolic hallmarks of parent tumors and SDH deficiency. We further demonstrate that temozolomide elicits DNA damage and apoptosis in our mSDH GIST models. Translating our in vitro discovery to the clinic, a cohort of patients with SDH-mutant GIST treated with temozolomide (n = 5) demonstrated a 40% objective response rate and 100% disease control rate, suggesting that temozolomide represents a promising therapy for this subset of GIST. Conclusions: We report the first methods to establish patient-derived mSDH tumor models, which can be readily employed for understanding patient-specific tumor biology and treatment strategies. We also demonstrate that temozolomide is effective in patients with mSDH GIST who are refractory to existing chemotherapeutic drugs (namely, TKIs) in clinic for GISTs, bringing a promising treatment option for these patients to clinic. See related commentary by Blakely et al., p. 3
- Published
- 2022
10. Tumor mutational burden and purity adjustment before and after treatment with temozolomide in 27 paired samples of glioblastoma: a prospective study
- Author
-
Joachim Weischenfeldt, Hans Skovgaard Poulsen, Petra Hamerlik, Olga Østrup, Christina Westmose Yde, Aidan Flynn, Jannick Brennum, Dorte Schou Nørøxe, Finn Cilius Nielsen, Ulrik Lassen, and Jane Skjøth-Rasmussen
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,tumor purity ,tumor mutational burden ,medicine.medical_treatment ,temozolomide ,Internal medicine ,immune therapy ,Biomarkers, Tumor ,Genetics ,medicine ,Humans ,Clinical significance ,Prospective Studies ,Prospective cohort study ,paired samples ,Research Articles ,RC254-282 ,Mutation Spectra ,Temozolomide ,business.industry ,glioblastoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,General Medicine ,Immune checkpoint ,Tumor Burden ,Radiation therapy ,Isocitrate dehydrogenase ,CpG site ,Mutation ,Molecular Medicine ,Neoplasm Recurrence, Local ,business ,Research Article ,medicine.drug - Abstract
Treatment of glioblastoma (GBM) remains a challenging task, with limited treatment options, none offering a cure. Immune therapy has proven effective across different cancers with remarkable response rates. Tumor mutational burden (TMB) is a marker of response, but technical and methodological differences in TMB estimates have made a proper assessment and comparison challenging. Here, we analyzed a prospective collection of paired samples from 35 patients with newly diagnosed GBM, all of whom were wild‐type (WT) for isocitrate dehydrogenase, before and after treatment with radiotherapy and temozolomide. Seven patients (20%) had O6‐methylguanine‐DNA methyltransferase‐methylated tumors. Six patients (17%) had two relapse surgeries, and tissue from all three surgeries was collected. We found that accurate evaluation of TMB was confounded by high variability in the cancer cell fraction of relapse samples. To ameliorate this, we developed a model to adjust for tumor purity based on the relative density distribution of variant allele frequencies in each primary–relapse pair. Additionally, we examined the mutation spectra of shared and private mutations. After tumor purity adjustment, we found TMB comparison reliable in tumors with tumor purity between 15% and 40%, resulting in 27/35 patients (77.1%). TMB remained unchanged from 0.65 mutations per megabase (Mb) to 0.67/Mb before and after treatment, respectively. Examination of the mutation spectra revealed a dominance of C > T transitions at CpG sites in both shared and relapse‐private mutations, consistent with cytosine deamination and the clock‐like mutational signature 1. We present and apply a cellularity correction approach that enables more accurate assessment of TMB in paired tumor samples. We did not find a significant increase in TMB after correcting for cancer cell fraction. Our study raises significant concerns when determining TMB. Although a small sample size, corrected TMB can have a clinical significance when stratifying patients to experimental treatment, for example, immune checkpoint therapy., Tumor mutational burden (TMB) is an important clinical marker used to potentially allocate patients for immune therapy. We estimated TMB before and after treatment in 35 paired samples from patients with glioblastoma. Surprisingly, relapse samples presented with a low degree of tumor purity, making intersampling comparison of TMB unreliable. We therefore developed a computational method to adjust for tumor purity in exome sequencing data and found comparison reliable in 27 paired samples. The underexplored role of tumor purity should be included in the development of a standardized method for TMB evaluation in order to stratify clinical data analysis and the evaluation of cancer treatment strategies.
- Published
- 2022
11. Efficacy of depatuxizumab mafodotin (ABT-414) monotherapy in patients with EGFR-amplified, recurrent glioblastoma: results from a multi-center, international study
- Author
-
Louis B. Nabors, David A. Reardon, John Simes, Lisa Roberts-Rapp, Ho-Jin Lee, Nicholas Butowski, Tobias Walbert, Andrew B. Lassman, Helen Wheeler, Priya Kumthekar, Andrew M. Scott, Peter Ansell, Tom Mikkelsen, Marta Penas-Prado, Kyriakos P. Papadopoulos, Martin J. van den Bent, Earle Bain, Zarnie Lwin, Erica Gomez, David Maag, Ryan Merrell, Hui K Gan, Hao Xiong, Kyle D. Holen, and Neurology
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Immunoconjugates ,EGFR ,Population ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Toxicology ,Depatuxizumab mafodotin ,03 medical and health sciences ,0302 clinical medicine ,Gefitinib ,Internal medicine ,Recurrent glioblastoma ,Humans ,Medicine ,Pharmacology (medical) ,Progression-free survival ,education ,Survival rate ,Aged ,Uncategorized ,Aged, 80 and over ,education.field_of_study ,Temozolomide ,business.industry ,Antimicrotubule agent ,Middle Aged ,ABT-414 ,ErbB Receptors ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Original Article ,Erlotinib ,Glioblastoma ,business ,Antibody–drug conjugate ,medicine.drug - Abstract
Purpose Patients with recurrent glioblastoma (rGBM) have a poor prognosis. Epidermal growth factor receptor (EGFR) gene amplification is present in ~ 50% of glioblastomas (GBMs). Depatuxizumab mafodotin (depatux-m), formerly ABT-414, is an antibody–drug conjugate that preferentially binds cells with EGFR amplification, is internalized and releases a potent antimicrotubule agent, monomethyl auristatin F (MMAF). Here we report the safety, pharmacokinetics, and efficacy of depatux-m monotherapy at the recommended Phase 2 dose (RPTD) in patients with EGFR-amplified, rGBM. Methods M12-356 (NCT01800695) is an open-label study with three escalation and expansion cohorts. Sixty-six patients with EGFR-amplified, rGBM were treated with depatux-m monotherapy at 1.25 mg/kg intravenously every 2 weeks. Adults with measurable rGBM, who were bevacizumab-naïve, with EGFR amplification were eligible. Results Among 66 patients, median age was 58 years (range 35–80). All patients were previously treated with radiotherapy/temozolomide. The most common adverse events (AEs) were eye related (91%), including blurred vision (65%), dry eye (29%), keratitis, and photophobia (27% each). Grade 3/4 AEs occurred in 42% of all patients, and ocular Grade 3/4 AEs occurred in 33% of patients overall. One patient (2%) had a Grade 4 ocular AE. Ocular AEs were manageable and usually resolved once treatment with depatux-m ceased. The objective response rate was 6.8%, the 6-month progression-free survival rate was 28.8%, and the 6-month overall survival rate was 72.5%. Conclusion Depatux-m monotherapy displayed frequent but mostly Grade 1/2 ocular toxicities. A PFS6 of 28.8% was observed in this rGBM population, warranting further study. Electronic supplementary material The online version of this article (doi:10.1007/s00280-017-3451-1) contains supplementary material, which is available to authorized users.
- Published
- 2023
- Full Text
- View/download PDF
12. Glioblastoma Cell–Derived lncRNA-Containing Exosomes Induce Microglia to Produce Complement C5, Promoting Chemotherapy Resistance
- Author
-
Shupeng Li, Jinquan Cai, Changxiao Yang, Jie Qin, Nan Sun, Ziwei Li, Kaifu Tian, Chuanlu Jiang, Jianguang Ming, Pengfei Wu, Caijun Zha, Bo Han, Lulu Li, Yifei Song, Xiangqi Meng, Tengfei Qi, Yangong Zhang, Pandeng Wang, and Lejia Ren
- Subjects
Cancer Research ,Combination therapy ,DNA damage ,medicine.medical_treatment ,Immunology ,Exosomes ,Transfection ,Mice ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Complement component 5 ,Tumor microenvironment ,Temozolomide ,Microglia ,business.industry ,Complement C5 ,Immunotherapy ,Xenograft Model Antitumor Assays ,Microvesicles ,Disease Models, Animal ,medicine.anatomical_structure ,Drug Resistance, Neoplasm ,Cancer research ,RNA, Long Noncoding ,Glioblastoma ,business ,medicine.drug - Abstract
Glioblastoma (GBM), the most common malignant primary brain cancer in adults, nearly always becomes resistant to current treatments, including the chemotherapeutic temozolomide (TMZ). The long noncoding RNA (lncRNA) TMZ-associated lncRNA in GBM recurrence (lnc-TALC) promotes GBM resistance to TMZ. Exosomes can release biochemical cargo into the tumor microenvironment (TME) or transfer their contents, including lncRNAs, to other cells as a form of intercellular communication. In this study, we found that lnc-TALC could be incorporated into exosomes and transmitted to tumor-associated macrophages (TAM) and could promote M2 polarization of the microglia. This M2 polarization correlated with secretion of the complement components C5/C5a, which occurred downstream of lnc-TALC binding to ENO1 to promote the phosphorylation of p38 MAPK. In addition, C5 promoted the repair of TMZ-induced DNA damage, leading to chemotherapy resistance, and C5a-targeted immunotherapy showed improved efficacy that limited lnc-TALC–mediated TMZ resistance. Our results reveal that exosome-transmitted lnc-TALC could remodel the GBM microenvironment and reduce tumor sensitivity to TMZ chemotherapy, indicating that the lnc-TALC–mediated cross-talk between GBM cells and microglia could attenuate chemotherapy efficacy and pointing to potential combination therapy strategies to overcome TMZ resistance in GBM. See related Spotlight by Zhao and Xie, p. 1372.
- Published
- 2021
13. Pediatric glioblastoma treated with concurrent radiotherapy and adjuvant Temozolomide: A case study of the treatment of pediatric GBM by the technique of volumetric modulated arc therapy
- Author
-
Nusret Salkica, Mirjana Ristanić, Halil Ćorović, Lejla Altumbabić, Velda Smajlbegović, and Enis Tinjak
- Subjects
Oncology ,medicine.medical_specialty ,Temozolomide ,Pediatric glioblastoma ,business.industry ,medicine.medical_treatment ,Volumetric modulated arc therapy ,Radiation therapy ,Internal medicine ,medicine ,General Earth and Planetary Sciences ,business ,Adjuvant ,General Environmental Science ,medicine.drug - Abstract
Introduction: Glioblastoma in children (pGBM) occurs somewhat less frequently than in adults. Pediatric pGBMs have a different molecular profile than GBM for adults. The aim of the presentation of this case is the possibility of the effectiveness of the GBM radiation method and the evaluation of magnetic resonance imaging, and the monitoring of the treatment outcome of the patient.Material and methods: The case study is of the retrospective-prospective type. Medical documentation, magnetic resonance imaging, and chronologically monitored evaluation of the findings from November 2018 to August 2021 were used to present the study. The postoperative course was analyzed, as well as the effect of concurrent chemoradiotherapy, VMAT radiotherapy and adjuvant chemotherapy with Temozolomide in a patient aged 4 years and 6 months, comparing treatment outcome with median and overall survival in glioblastoma.Results: The pediatric patient after being diagnosed with high-grade glioma in 2018 is so far in very good general condition, without signs of physical and psycho-social defects, which compared to the scientifically proven median of survival indicates a good therapeutic effect. Volumetrically modulated arc radiotherapy with the use of modern IGRT verification techniques and with the use of chemotherapy with Temozoloimod, has proven to be a still effective oncological method treatment of GBM. For the final outcome of the disease and the effect of therapeutic modalities, the patient's condition and evaluation of magnetic resonance imaging will be monitored. The result supports further research into this therapeutic regimen.Conclusion: Glioblastoma is a very aggressive tumor, which occurs somewhat less frequently in the pediatric population than in adults, but is a very fatal disease. Surgical resection followed by concurrent chemoradiotherapy, with adjuvant Temozolomide is still the method of choice in the treatment of glioblastoma.
- Published
- 2021
14. Temporal Trends in Glioblastoma Survival
- Author
-
Michael W. Ruff, Derek R. Johnson, Bryan J. Neth, and Ivan D. Carabenciov
- Subjects
Adult ,Oncology ,medicine.medical_specialty ,Temozolomide ,Bevacizumab ,Brain Neoplasms ,business.industry ,Proportional hazards model ,Improved survival ,Kaplan-Meier Estimate ,medicine.disease ,Confidence interval ,Internal medicine ,Epidemiology ,medicine ,Humans ,Glioblastoma ,business ,Survival analysis ,medicine.drug - Abstract
BACKGROUND Survival of patients with glioblastoma (GBM) increased in the 2000s, most prominently after the addition of temozolomide to the standard-of-care treatment protocol. The reason for subsequent improvements in survival in the late 2000s and early 2010s was less clear, with explanations including the introduction of bevacizumab, better surgical methods, and advances in supportive care. It is uncertain whether the trend of improving population-level survival has continued. MATERIALS AND METHODS Data from the Surveillance, Epidemiology, and End Results (SEER) Program was analyzed comparing survival of adult GBM patients diagnosed in consecutive 3-year periods from 2000 to 2017. Kaplan-Meier survival analysis and Cox proportional hazards models were used. RESULTS A total of 38,352 patients diagnosed with GBM between 2000 and 2017 met inclusion criteria. Median survival and percent survival to 12 and 24 months all progressively increased between 2000 and 2011. There were no significant differences in survival comparing 2009-2011 with 2012-2014 or 2015-2017. During the 2015-2017 period, median survival was 11 months, with 12 and 24-month survival proportions of 45.7% (95% confidence interval, 44.5-47.0) and 19.0% (95% confidence interval, 18.6-21.2), respectively. CONCLUSIONS After a period of progressive improvement in GBM survival between 2000 and 2011, survival plateaued. Subsequent advances since 2011 have not yet been translated to improved survival on the population-level as of 2017.
- Published
- 2021
15. Anlotinib Induces a T Cell–Inflamed Tumor Microenvironment by Facilitating Vessel Normalization and Enhances the Efficacy of PD-1 Checkpoint Blockade in Neuroblastoma
- Author
-
Qiang Zhao, Yao Lu, Bingying Luo, Jie Yin, Daowei Wang, Peng Chen, Jie Yan, Long Li, Yangyang Wang, Jian Zheng, Jun Xiao, Zhenyi Xue, and Yudong Su
- Subjects
Cancer Research ,Indoles ,medicine.drug_class ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Programmed Cell Death 1 Receptor ,Tyrosine-kinase inhibitor ,Flow cytometry ,Mice ,Neuroblastoma ,Cell Line, Tumor ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Tumor microenvironment ,Temozolomide ,medicine.diagnostic_test ,business.industry ,Immunosuppression ,medicine.disease ,Irinotecan ,medicine.anatomical_structure ,Oncology ,Quinolines ,Cancer research ,Neoplasm Recurrence, Local ,business ,medicine.drug - Abstract
Purpose: Anlotinib has achieved good results in clinical trials of a variety of cancers. However, the effects of anlotinib on the tumor microenvironment (TME) and systemic immunity have not been reported. There is an urgent need to identify the underlying mechanism to reveal new opportunities for its application in neuroblastoma (NB) and other cancers. Understanding the mechanism will hopefully achieve the goal of using the same method to treat different cancers. Experimental Design: This study used bioinformatics, NB syngeneic mouse models, flow cytometry, RNA-seq, and immunofluorescence staining to explore the mechanisms of anlotinib on the TME, and further explored anlotinib-containing combination treatment strategies. Results: We proved that anlotinib facilitates tumor vessel normalization at least partially through CD4+ T cells, reprograms the immunosuppressive TME into an immunostimulatory TME, significantly inhibits tumor growth, and effectively prevents systemic immunosuppression. Moreover, the combination of anlotinib with a PD-1 checkpoint inhibitor counteracts the immunosuppression caused by the upregulation of PD-L1 after monotherapy, extends the period of vascular normalization, and finally induces NB regression. Conclusions: To our knowledge, this study is the first to dynamically evaluate the effect of a multitarget antiangiogenic tyrosine kinase inhibitor on the TME. These findings have very important clinical value in guiding the testing of related drugs in NB and other cancers. Based on these findings, we are conducting a phase II clinical study (NCT04842526) on the efficacy and safety of anlotinib, irinotecan, and temozolomide in the treatment of refractory or relapsed NB, and hopefully we will observe patient benefit.
- Published
- 2021
16. Hematological adverse events in the management of glioblastoma
- Author
-
Rachael M. Morgan, Zin W. Myint, Rani Jayswal, Heidi L. Weiss, Allison R Butts, John L. Villano, Catherine R Garcia, and Chi Wang
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,medicine.medical_treatment ,Article ,Internal medicine ,Temozolomide ,medicine ,Adjuvant therapy ,Humans ,Progression-free survival ,Adverse effect ,Antineoplastic Agents, Alkylating ,Randomized Controlled Trials as Topic ,Leukopenia ,Brain Neoplasms ,business.industry ,Thrombocytopenia ,Regimen ,Neurology ,Neurology (clinical) ,medicine.symptom ,Glioblastoma ,business ,Adjuvant ,medicine.drug - Abstract
Background Hematological adverse events (HAEs) are common during treatment for glioblastoma (GBM), usually associated with temozolomide (TMZ). Their clinical value is uncertain, as few investigations have focused on outcomes for HAEs during GBM treatment. Methods We combined data from two randomized clinical trials, RTOG 0525 and RTOG 0825, to analyze HAEs during treatment for GBM. We investigated differences between chemoradiation and adjuvant therapy, and by regimen received during adjuvant treatment. Results 1454 patients participated in these trials, of which 1154 (79.4%) developed HAEs. During chemoradiation, 44.4% of patients developed HAEs (54% involving more than one cell line), and were most commonly lymphopenia (50.6%), and thrombocytopenia (47.5%). During adjuvant treatment, 45% of patients presented HAEs (78.6% involving more than one cell line), and were more commonly leukopenia (62.7%), and thrombocytopenia (62.3%). Median overall survival (OS) and progression free survival (PFS) were longer in patients with HAEs (OS 19.4 months and PFS 9.9 months) compared to those with other or no adverse events (OS 14.1 months and PFS 5.9 months). There was no significant difference in survival between grade 1 and/or 2 versus grade 3 and/or 4 HAEs. History of HAEs during chemoradiation was a protective factor for presentation of HAEs during adjuvant therapy. Conclusion HAEs are common during GBM treatment, and often involve more than one cell line (more likely during adjuvant therapy). HAEs may be associated with prolonged OS and PFS, particularly during adjuvant therapy. HAEs during chemoradiation was a protective factor for HAEs during adjuvant therapy.
- Published
- 2021
17. Stereotactic radiosurgery for IDH wild type glioblastoma: an international, multicenter study
- Author
-
Dev N Patel, Manjul Tripathi, Erik P. Sulman, David E. Arsanious, Ronald E Warnick, Gene Barnett, Joshua S Weir, Violaine Delabar, Angel Mota, Douglas Kondziolka, Ghusn Al Sideiri, Christopher P. Cifarelli, Adomas Bunevicius, Huai-Che Yang, Narendra Kumar, David Mathieu, Jason P. Sheehan, Kenneth Bernstein, Cheng-Chia Lee, Farid El Hefnawi, Stylianos Pikis, Basem Dahshan, Yavuz Samanci, Selçuk Peker, and Herwin Speckter
- Subjects
Oncology ,Cancer Research ,Chemotherapy ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,medicine.disease ,Radiosurgery ,Isocitrate dehydrogenase ,Neurology ,Multicenter study ,Tumor progression ,Internal medicine ,parasitic diseases ,medicine ,Neurology (clinical) ,Progression-free survival ,business ,medicine.drug ,Glioblastoma - Abstract
Objective Isocitrate dehydrogenase (IDH) mutation status is recommended used for diagnosis and prognostication of glioblastoma patients. We studied efficacy and safety of stereotactic radiosurgery (SRS) for patients with recurrent IDH-wt glioblastoma. Methods Consecutive patients treated with SRS for IDH-wt glioblastoma were pooled for this retrospective observational international multi-institutional study from institutions participating in the International Radiosurgery Research Foundation. Results Sixty patients (median age 61 years) underwent SRS (median dose 15 Gy and median treatment volume: 7.01 cm3) for IDH-wt glioblastoma. All patients had histories of surgery and chemotherapy with temozolomide, and 98% underwent fractionated radiation therapy. MGMT status was available for 42 patients, of which half of patients had MGMT mutant glioblastomas. During median post-SRS imaging follow-up of 6 months, 52% of patients experienced tumor progression. Median post-SRS progression free survival was 4 months. SRS prescription dose of > 14 Gy predicted longer progression free survival [HR 0.357 95% (0.164-0.777) p = 0.009]. Fifty-percent of patients died during post-SRS clinical follow-up that ranged from 1 to 33 months. SRS treatment volume of > 5 cc emerged as an independent predictor of shorter post-SRS overall survival [HR 2.802 95% CI (1.219-6.444) p = 0.02]. Adverse radiation events (ARE) suggestive of radiation necrosis were diagnosed in 6/55 (10%) patients and were managed conservatively in the majority of patients. Conclusions SRS prescription dose of > 14 Gy is associated with longer progression free survival while tumor volume of > 5 cc is associated with shorter overall survival after SRS for IDH-wt glioblastomas. AREs are rare and are typically managed conservatively.
- Published
- 2021
18. Selected cytostatic drugs in the treatment of brain glioma – literature review
- Author
-
Natalia Walo, Kinga Mitruczuk, Jarosław Bała, and Paula Wróblewska-Łuczka
- Subjects
Vincristine ,Brain glioma ,Temozolomide ,Bevacizumab ,business.industry ,Lomustine ,medicine.disease ,Procarbazine ,Glioma ,medicine ,Cytostatic drugs ,Cancer research ,General Earth and Planetary Sciences ,business ,General Environmental Science ,medicine.drug - Published
- 2021
19. Rh-endostatin combined with chemotherapy in patients with advanced or recurrent mucosal melanoma: retrospective analysis of real-world data
- Author
-
Yunyi Kong, Yanchun Meng, Shiyu Jiang, Xin Liu, Jun Cao, Feng Jin, ChunmengWang, Xiaowei Zhang, Zhiguo Luo, Yu Xu, Yong Chen, and Huijuan Yang
- Subjects
Proto-Oncogene Proteins B-raf ,Neuroblastoma RAS viral oncogene homolog ,China ,medicine.medical_specialty ,Skin Neoplasms ,Dacarbazine ,medicine.medical_treatment ,Population ,Gastroenterology ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Pharmacology (medical) ,education ,Melanoma ,Retrospective Studies ,Pharmacology ,Chemotherapy ,education.field_of_study ,Temozolomide ,business.industry ,Standard treatment ,Mucosal melanoma ,Cancer ,medicine.disease ,Endostatins ,Oncology ,Cisplatin ,business ,medicine.drug - Abstract
Background Mucosal melanoma is rare and has distinct clinical and genetic features. Even with advances in targeted and immune therapies, the survival of patients with advanced or recurrent mucosal melanomas remains poor. The standard treatment remains controversial and we conducted this real-world study aimed to explore continuous intravenous recombinant human endostatin (Rh-endostatin) infusion plus chemotherapy in this population in the first-line setting. Methods Overall, 43 patients with advanced or recurrent mucosal melanoma treated at Fudan University Shanghai Cancer Center between April 2017 and August 2020 were retrospectively included. Patients received dacarbazine plus cisplatin or temozolomide plus cisplatin per the investigators' preference. Rh-endostatin (105 mg/m2) was administered with continuous infusion for 168 h (Civ 168 h). Results Of the 43 patients, 72.1% had metastatic disease, and the most common primary site was the gastrointestinal tract (51.2%). The most commonly observed mutations were NRAS (23.1%), BRAF (7.7%) and CKIT mutations (5.1%). An objective response was observed in 12 (30.0%) of the 40 evaluable patients, and disease control was achieved in 31 (77.5%) patients. With a median follow-up of 17.6 months, the median progression-free survival (PFS) and overall survival (OS) were 4.9 and 15.3 months, respectively. Additionally, high lymphocyte-to-monocyte ratio (LMR) (p = 0.023, HR 0.29, 95% CI: 0.10-0.84) and BRAF/KIT/RAS mutation (p = 0.028, HR 0.24, 95% CI: 0.07-0.86) were independently correlated with prolonged OS. Toxicity was manageable overall. Conclusion Continuous Rh-endostatin infusion plus chemotherapy was effective and safe for the treatment of advanced or recurrent mucosal melanoma. High LMR was correlated with favorable PFS and OS in this patient population.
- Published
- 2021
20. Improving temozolomide biopharmaceutical properties in glioblastoma multiforme (GBM) treatment using GBM-targeting nanocarriers
- Author
-
Juliana Hofstatter Azambuja, Jessyca Aparecida Paes Dutra, Jonatas Lobato Duarte, Luiza Ribeiro Nicoleti, Sara Teresinha Olalla Saad, Leonardo Delello Di Filippo, Marlus Chorilli, Marcela Tavares Luiz, Universidade Estadual Paulista (UNESP), Universidade Estadual de Campinas (UNICAMP), and Universidade de São Paulo (USP)
- Subjects
Drug ,Cancer therapy ,medicine.medical_treatment ,media_common.quotation_subject ,Pharmaceutical Science ,Context (language use) ,Drug resistance ,Brain cancer ,urologic and male genital diseases ,Drug Delivery Systems ,Temozolomide ,medicine ,Nanotechnology ,Animals ,Humans ,Functionalization ,Antineoplastic Agents, Alkylating ,media_common ,Drug Carriers ,Chemotherapy ,Tumor microenvironment ,Brain Neoplasms ,urogenital system ,business.industry ,General Medicine ,female genital diseases and pregnancy complications ,nervous system diseases ,Biopharmaceutical ,Drug Resistance, Neoplasm ,Cancer research ,Nanoparticles ,Nanocarriers ,Glioblastoma ,business ,Biotechnology ,medicine.drug - Abstract
Made available in DSpace on 2022-05-01T08:44:46Z (GMT). No. of bitstreams: 0 Previous issue date: 2021-11-01 Glioblastoma multiforme (GBM) is the most common primary brain cancer. GBM has aggressive development, and the pharmacological treatment remains a challenge due to GBM anatomical characteristics’ (the blood–brain barrier and tumor microenvironment) and the increasing resistance to marketed drugs, such as temozolomide (TMZ), the first-line drug for GBM treatment. Due to physical–chemical properties such as short half-life time and the increasing resistance shown by GBM cells, high doses and repeated administrations are necessary, leading to significant adverse events. This review will discuss the main molecular mechanisms of TMZ resistance and the use of functionalized nanocarriers as an efficient and safe strategy for TMZ delivery. GBM-targeting nanocarriers are an important tool for the treatment of GBM, demonstrating to improve the biopharmaceutical properties of TMZ and repurpose its use in anti-GBM therapy. Technical aspects of nanocarriers will be discussed, and biological models highlighting the advantages and effects of functionalization strategies in TMZ anti-GBM activity. Finally, conclusions regarding the main findings will be made in the context of new perspectives for the treatment of GBM using TMZ as a chemotherapy agent, improving the sensibility and biological anti-tumor effect of TMZ through functionalization strategies. School of Pharmaceutical Sciences São Paulo State University (UNESP) Hematology and Transfusion Medicine Center University of Campinas (UNICAMP) School of Pharmaceutical Science of Ribeirão Preto University of São Paulo (USP) School of Pharmaceutical Sciences São Paulo State University (UNESP)
- Published
- 2021
21. Effect of long-term adjuvant temozolomide chemotherapy on primary glioblastoma patient survival
- Author
-
Bin Huang, Zuan Yu, and Risheng Liang
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Single Center ,Long-term adjuvant chemotherapy ,progression-freesurvival ,Internal medicine ,medicine ,Temozolomide ,Humans ,RC346-429 ,Survival rate ,Antineoplastic Agents, Alkylating ,Retrospective Studies ,Chemotherapy ,business.industry ,Proportional hazards model ,Brain Neoplasms ,Standard treatment ,Research ,General Medicine ,Cancer survival ,Dacarbazine ,Regimen ,Chemotherapy, Adjuvant ,Quality of Life ,Neurology (clinical) ,Neurology. Diseases of the nervous system ,business ,Glioblastoma ,Adjuvant ,medicine.drug - Abstract
Objective Glioblastoma multiforme (GBM) is the most common primary malignant central nervous system (CNS) tumor. The Stupp regimen is the standard treatment, although the optimal number of temozolomide (TMZ) treatment cycles remains controversial. We compared the effects of standard 6 cycles versus > 6 cycles of TMZ chemotherapy post-surgery with concurrent chemoradiotherapy on primary GBM patient survival. Patients and methods We performed a single center retrospective study of GBM patients that underwent total resection, concurrent chemoradiotherapy, and at least 6 cycles of adjuvant TMZ chemotherapy from June 2011 to August 2018. Patients were divided into 2 groups based on adjuvant TMZ treatment plan: Group A(n = 27): standard 6-cycle adjuvant TMZ therapy and Group B(n = 26): > 6 cycles of adjuvant TMZ therapy. Primary endpoints were progression-free survival (PFS) and overall survival (OS). Continuous variables were analyzed by ANOVA, and the Kaplan-Meier method was used to evaluate PFS and OS. Univariate and multivariate COX analyses determined correlation between survival rates and covariates. We used The Mini Mental State Examination (MMSE) and Karnofsky Performance Status (KPS) to assess patients’ neurocognitive function and quality of life. Results After follow-up, median PFS was 15 months in in Group A (95%CI 9.5–20.5) and 20.1 months in Group B (95%CI 15.9–24.4). Group A median OS was 19.4 months (95%CI 15.5–23.2), compared to 25.6 months in Group B (95%CI 20.4–30.8). The 2-year survival rate of Groups A and B was 36% was 66%, respectively (P = 0.02). and 5-year survival was 7% in both. Multivariate COX regression analysis showed association between patient PFS and long-period adjuvant chemotherapy, but not OS. There were no significant difference in disability or quality of life during treatment with Stupp protocol, but differences in MMSE and KPS were in favour of the Groups B after year 1 of the treatment (P Conclusions Long-term adjuvant TMZ chemotherapy was beneficial for PFS and 2-year survival rate in GBM patients, and improved their quality of life contemporarily. But OS was not significantly improved.
- Published
- 2021
22. Treatment of anaplastic gliomas
- Author
-
Martin J. van den Bent, Marjolein Geurts, Tom J. Snijders, Neurology, and Erasmus MC other
- Subjects
Oncology ,Cancer Research ,Vincristine ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,Retrospective cohort study ,medicine.disease ,Chemotherapy regimen ,Clinical trial ,Isocitrate dehydrogenase ,SDG 3 - Good Health and Well-being ,Internal medicine ,medicine ,business ,Adjuvant ,medicine.drug ,Anaplastic astrocytoma - Abstract
PURPOSE OF REVIEW: Evolving molecular data have led to a new and advanced grading system of anaplastic glioma. In everyday practice, physicians have to translate evidence from old clinical trials into evidence meeting the reclassified tumor types. RECENT FINDINGS: New biomarkers allow the identification of anaplastic glioma with relatively poor prognosis and with prognosis similar to glioblastoma. An update with molecular analysis of the phase 3 CATNON trial demonstrates the benefit of adjuvant temozolomide (TMZ) to be dependent on the mutational status of isocitrate dehydrogenase. In the ongoing debate on the optimal chemotherapy regimen, a large retrospective study suggesting a better tumor control with vincristine (PCV) as compared to TMZ is added to the evidence. The best timing for treatment of anaplastic astrocytoma also remains a matter of controversy. A recent study shows that even in selected patients with anaplastic glioma with foci of malignant tumor following (sub)total resection, postponement of medical treatment can be considered. SUMMARY: In clinical practice, the trade-off between efficacy and (acute and long-term) toxicity of treatments needs to be re-evaluated for the newly (molecularly) defined entities. Updates from past clinical trials on anaplastic glioma with molecular analysis and subgroup analyses are needed to further guide treatment decisions.
- Published
- 2021
23. Afatinib and radiotherapy, with or without temozolomide, in patients with newly diagnosed glioblastoma: results of a phase I trial
- Author
-
Karine Pemberton, Fiona Harris, Jennifer Schaible, Frank Saran, Sarah Jefferies, Agnieszka Cseh, Michael Brada, Shaun Bender, Liam Welsh, Catherine McBain, Allan James, Rao Gattamaneni, Saran, Frank [0000-0002-3448-5064], and Apollo - University of Cambridge Repository
- Subjects
Oncology ,Adult ,Cancer Research ,medicine.medical_specialty ,Afatinib ,ErbB ,Internal medicine ,Glioma ,Dose-escalation ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Temozolomide ,Humans ,Epidermal growth factor receptor ,biology ,Radiotherapy ,business.industry ,Brain Neoplasms ,medicine.disease ,Dacarbazine ,Regimen ,Treatment Outcome ,Neurology ,Concomitant ,biology.protein ,Clinical Study ,Neurology (clinical) ,business ,Glioblastoma ,medicine.drug - Abstract
Funder: Boehringer Ingelheim; doi: http://dx.doi.org/10.13039/100001003, BACKGROUND: Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor in adults. Amplification or overexpression of the epidermal growth factor receptor gene, part of the ErbB family, occur in approximately 40% and 60% of patients with GBM, respectively. We present data from a dose-finding study of the ErbB inhibitor afatinib in combination with radiotherapy (RT), with or without temozolomide (TMZ), in patients with GBM. METHODS: This was a phase I, open-label, 3 + 3 dose-escalation trial in patients with newly-diagnosed, histologically-confirmed grade 4 malignant glioma and proven O6-methylguanine-DNA methyltransferase gene promoter methylation status. The primary endpoint was the maximum tolerated dose (MTD) of continuous daily afatinib when given in combination with RT, with (regimen M) or without (regimen U) concomitant TMZ treatment. RESULTS: Fifty-five patients were enrolled; 36 received ≥ 1 dose of trial medication (regimen M, n = 20, regimen U, n = 16). Afatinib was discontinued by all patients during the study. Reasons for afatinib discontinuation (regimen M/U) included disease progression (45%/50%), dose-limiting toxicity (10%/0%), and other adverse events (AEs; 35%/38%). The most frequently reported AEs with either regimen were diarrhea and rash, with no new safety signals identified. The MTD was determined as afatinib 30 mg in combination with daily TMZ and RT, and afatinib 40 mg in combination with RT alone. CONCLUSIONS: This study identified the MTD for afatinib in combination with RT, with and without TMZ, in patients with GBM. Further studies of afatinib in patients with GBM are warranted and should be based on appropriate biomarker-based preselection. TRIAL REGISTRATION: NCT00977431 (first posted September 15, 2009).
- Published
- 2021
24. Primary vitreoretinal lymphoma: a diagnostic and management challenge
- Author
-
Denis Malaise, Nathalie Cassoux, and Carole Soussain
- Subjects
Oncology ,medicine.medical_specialty ,Retinal Neoplasms ,medicine.medical_treatment ,Immunology ,Biochemistry ,Retina ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,Humans ,Lenalidomide ,Chemotherapy ,Temozolomide ,business.industry ,Disease Management ,Consolidation Chemotherapy ,Cell Biology ,Hematology ,medicine.disease ,Lymphoma ,Vitreous Body ,Transplantation ,chemistry ,Ibrutinib ,Rituximab ,business ,medicine.drug - Abstract
Primary vitreoretinal lymphoma (PVRL) is a rare form of primary central nervous system (CNS) lymphoma (PCNSL) arising in the intraocular compartment without brain involvement. Despite its apparent indolent clinical course, PVRL can cause permanent vision loss and CNS relapse, the major cause of death in patients with PVRL. The pathophysiology of PVRL is unknown. As in PCNSL, the transformation of the tumor cells likely originates outside the CNS, before the cells migrate to the eye and proliferate within an immune-permissive microenvironment. PVRL exhibits a biased immunoglobulin repertoire, suggesting underlying antigen selection. The diagnosis remains challenging, requiring close coordination between ophthalmologists and cytologists. Because of their rarity and fragility in the vitreous, lymphoma cells cannot always be identified. Interleukin levels, molecular biology, and imaging are used in combination with clinical ophthalmological examination to support the diagnosis of PVRL. Multi-institutional prospective studies are urgently needed to validate the equivocal conclusions regarding treatments drawn from heterogeneous retrospective or small cohort studies. Intravitreal injection of methotrexate or rituximab or local radiotherapy is effective at clearing tumor cells within the eyes but does not prevent CNS relapse. Systemic treatment based on high-dose methotrexate chemotherapy, with or without local treatment, might reduce this risk. At relapse, intensive consolidation chemotherapy followed by stem cell transplantation can be considered. Single-agent ibrutinib, lenalidomide, and temozolomide treatments are effective in patients with relapsed PVRL and should be tested as first-line treatments. Therapeutic response assessment based on clinical examination is improved by measuring cytokine levels but still needs to be refined.
- Published
- 2021
25. A multi-center prospective study of re-irradiation with bevacizumab and temozolomide in patients with bevacizumab refractory recurrent high-grade gliomas
- Author
-
Angela J. Fought, Karan Dixit, Vinai Gondi, Tim J. Kruser, Jeffrey Raizer, Minesh P. Mehta, Rimas V. Lukas, Sean Sachdev, Priya Kumthekar, Sean Grimm, Christina Amidei, Steven J. Chmura, and Martin K. Nicholas
- Subjects
Oncology ,Re-Irradiation ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,genetic structures ,Bevacizumab ,business.industry ,medicine.medical_treatment ,Recurrent Glioma ,medicine.disease ,eye diseases ,Radiation therapy ,Regimen ,Neurology ,Refractory ,Internal medicine ,Glioma ,medicine ,sense organs ,Neurology (clinical) ,business ,neoplasms ,medicine.drug - Abstract
Survival is dismal for bevacizumab refractory high-grade glioma patients. We prospectively investigated the efficacy of re-irradiation, bevacizumab, and temozolomide in bevacizumab-naive and bevacizumab-exposed recurrent high-grade glioma, without volume limitations, in a single arm trial. Recurrent high-grade glioma patients were stratified based on WHO grade (4 vs. grade 3 lymphopenia and 2 with > grade 3 thrombocytopenia. No radiographic or clinical radiation necrosis occurred. Re-irradiation with bevacizumab and temozolomide is a safe and feasible salvage treatment for patients with large volume bevacizumab-refractory high-grade glioma. Patients further from their initial radiotherapy may derive greater benefit with this regimen.
- Published
- 2021
26. Regulation of autophagy as a therapeutic option in glioblastoma
- Author
-
Swapan K. Ray and Amanda J Manea
- Subjects
Pharmacology ,Cancer Research ,Programmed cell death ,Temozolomide ,Combination therapy ,business.industry ,medicine.medical_treatment ,Biochemistry (medical) ,Clinical Biochemistry ,Autophagy ,Brain tumor ,Pharmaceutical Science ,Cellular homeostasis ,Cell Biology ,medicine.disease ,Radiation therapy ,Cancer research ,medicine ,Stem cell ,business ,neoplasms ,medicine.drug - Abstract
Around three out of one hundred thousand people are diagnosed with glioblastoma multiforme, simply called glioblastoma, which is the most common primary brain tumor in adults. With a dismal prognosis of a little over a year, receiving a glioblastoma diagnosis is oftentimes fatal. A major advancement in its treatment was made almost two decades ago when the alkylating chemotherapeutic agent temozolomide (TMZ) was combined with radiotherapy (RT). Little progress has been made since then. Therapies that focus on the modulation of autophagy, a key process that regulates cellular homeostasis, have been developed to curb the progression of glioblastoma. The dual role of autophagy (cell survival or cell death) in glioblastoma has led to the development of autophagy inhibitors and promoters that either work as monotherapies or as part of a combination therapy to induce cell death, cellular senescence, and counteract the ability of glioblastoma stem cells (GSCs) for initiating tumor recurrence. The myriad of cellular pathways that act upon the modulation of autophagy have created contention between two groups: those who use autophagy inhibition versus those who use promotion of autophagy to control glioblastoma growth. We discuss rationale for using current major therapeutics, their molecular mechanisms for modulation of autophagy in glioblastoma and GSCs, their potentials for making strides in combating glioblastoma progression, and their possible shortcomings. These shortcomings may fuel the innovation of novel delivery systems and therapies involving TMZ in conjunction with another agent to pave the way towards a new gold standard of glioblastoma treatment.
- Published
- 2021
27. Anti-invasive efficacy and survival benefit of the YAP-TEAD inhibitor verteporfin in preclinical glioblastoma models
- Author
-
Dominique Bozec, Halle Ronk, Raymund Yong, Joe Gerald Jesu Raj, Jane Houldsworth, Marc R. Birtwistle, Nadejda M. Tsankova, Meenakshi Mehrotra, Constantinos G. Hadjipanayis, Alexandros Bouras, Zarmeen Mussa, William Lam, Elena Zaslavsky, German Nudelman, Tanvi Joshi, and Anne Marie Barrette
- Subjects
Cancer Research ,Temozolomide ,business.industry ,Disease ,CDH2 ,medicine.disease ,Penetrance ,Verteporfin ,Metastasis ,Transcriptome ,Oncology ,medicine ,Cancer research ,Neurology (clinical) ,business ,TEAD1 ,medicine.drug - Abstract
Background Glioblastoma (GBM) remains a largely incurable disease as current therapy fails to target the invasive nature of glioma growth in disease progression and recurrence. Here, we use the FDA-approved drug and small molecule Hippo inhibitor Verteporfin (VP) to target YAP-TEAD activity, known to mediate convergent aspects of tumor invasion/metastasis, and assess the drug’s efficacy and survival benefit in GBM models. Methods Up to 8 low-passage patient-derived GBM cell lines with distinct genomic drivers, including 3 primary/recurrent pairs, were treated with VP or vehicle (VEH) to assess in vitro effects on proliferation, migration, invasion, YAP-TEAD activity, and transcriptomics. Patient-derived orthotopic xenograft (PDX) models were used to assess VP’s brain penetrance and effects on tumor burden and survival. Results VP treatment disturbed YAP/TAZ-TEAD activity; disrupted transcriptome signatures related to invasion, epithelial-to-mesenchymal, and proneural-to-mesenchymal transition, phenocopying TEAD1-knockout effects; and impaired tumor migration/invasion dynamics across primary and recurrent GBM lines. In an aggressive orthotopic PDX GBM model, short-term VP treatment consistently diminished core and infiltrative tumor burden, which was associated with decreased tumor expression of Ki67, nuclear YAP, TEAD1, and TEAD-associated targets EGFR, CDH2, and ITGB1. Finally, long-term VP treatment appeared nontoxic and conferred survival benefit compared to VEH in 2 PDX models: as monotherapy in primary (de novo) GBM and in combination with Temozolomide chemoradiation in recurrent GBM, where VP treatment associated with increased MGMT methylation. Conclusions We demonstrate combined anti-invasive and anti-proliferative efficacy for VP with survival benefit in preclinical GBM models, indicating potential therapeutic value of this already FDA-approved drug if repurposed for GBM patients.
- Published
- 2021
28. Znaczenie badań genetycznych i radiologicznych w diagnostyce i doborze terapii glejaków mózgu u dorosłych
- Author
-
Gabriela Janus-Szymańska, Łukasz Waszczuk, and Jagoda Jacków-Nowicka
- Subjects
Trametinib ,In vivo magnetic resonance spectroscopy ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,Bevacizumab ,business.industry ,Dabrafenib ,Oncology ,medicine ,Histopathology ,Radiology ,Differential diagnosis ,Vemurafenib ,business ,medicine.drug - Abstract
Molecular and imaging studies are applied along with histopathology in diagnosis and differential diagnosis of brain gliomas and they enable personalised clinical management. With knowledge of the patient’s clinical condition, a decision whether to observe the patient or proceed to immediate surgical treatment is made based on imaging results. On the other hand, knowledge of molecular predictive markers allows optimisation of chemotherapeutic decisions, e.g., introduction of personalised therapy (application of such drugs as temozolomide, bevacizumab, vemurafenib, dabrafenib and trametinib).
- Published
- 2021
29. ATR inhibition reverses the resistance of homologous recombination deficient MGMTlow/MMRproficient cancer cells to temozolomide
- Author
-
Michael E. Mullendore, Deborah Wilsker, Anne Monks, Melinda G. Hollingshead, Lara H. El Touny, Sergio Y. Alcoser, Angie B. Dull, Curtis Hose, Erik Harris, Michelle M. Gottholm-Ahalt, Annamaria Rapisarda, Beverly A. Teicher, Ralph E. Parchment, James H. Doroshow, and John Connelly
- Subjects
REV3L ,Temozolomide ,DNA damage ,business.industry ,RAD51 ,Cancer ,medicine.disease ,MMR ,chemistry.chemical_compound ,ATR ,Oncology ,chemistry ,In vivo ,HR ,Cancer cell ,medicine ,Cancer research ,Growth inhibition ,TMZ ,business ,Priority Research Paper ,medicine.drug - Abstract
The therapeutic efficacy of temozolomide (TMZ) is hindered by inherent and acquired resistance. Biomarkers such as MGMT expression and MMR proficiency are used as predictors of response. However, not all MGMTlow/−ve/MMRproficient patients benefit from TMZ treatment, indicating a need for additional patient selection criteria. We explored the role of ATR in mediating TMZ resistance and whether ATR inhibitors (ATRi) could reverse this resistance in multiple cancer lines. We observed that only 31% of MGMTlow/−ve/MMRproficient patient-derived and established cancer lines are sensitive to TMZ at clinically relevant concentrations. TMZ treatment resulted in DNA damage signaling in both sensitive and resistant lines, but prolonged G2/M arrest and cell death were exclusive to sensitive models. Inhibition of ATR but not ATM, sensitized the majority of resistant models to TMZ and resulted in measurable DNA damage and persistent growth inhibition. Also, compromised homologous recombination (HR) via RAD51 or BRCA1 loss only conferred sensitivity to TMZ when combined with an ATRi. Furthermore, low REV3L mRNA expression correlated with sensitivity to the TMZ and ATRi combination in vitro and in vivo. This suggests that HR defects and low REV3L levels could be useful selection criteria for enhanced clinical efficacy of an ATRi plus TMZ combination.
- Published
- 2021
30. Lomustine and nimustine exert efficient antitumor effects against glioblastoma models with acquired temozolomide resistance
- Author
-
Takashi Fujii, Daisuke Kawauchi, Eita Uchida, Atsuo Yoshino, Yoshitaka Narita, Nobuyoshi Sasaki, Kojiro Wada, Tatsuya Kobayashi, Masamichi Takahashi, Koichi Ichimura, Arata Tomiyama, Kaishi Satomi, Shun Yamamuro, Akihide Kondo, and Tomoyuki Nakano
- Subjects
Cancer Research ,Nitrosourea ,nitrosourea ,lomustine ,Mice, Nude ,Salvage therapy ,Antineoplastic Agents ,Methylation ,Histones ,Mice ,chemistry.chemical_compound ,In vivo ,Temozolomide ,medicine ,Animals ,Humans ,U87 ,nimustine ,DNA Modification Methylases ,neoplasms ,Salvage Therapy ,Mice, Inbred BALB C ,Brain Neoplasms ,business.industry ,Tumor Suppressor Proteins ,Nimustine ,glioblastoma ,temozolomide resistance ,Original Articles ,General Medicine ,Lomustine ,Xenograft Model Antitumor Assays ,nervous system diseases ,DNA Repair Enzymes ,Drug Discovery and Delivery ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,Apoptosis ,Cancer research ,Female ,Original Article ,Neoplasm Recurrence, Local ,business ,Injections, Intraperitoneal ,medicine.drug - Abstract
Glioblastomas (GBM) often acquire resistance against temozolomide (TMZ) after continuous treatment and recur as TMZ‐resistant GBM (TMZ‐R‐GBM). Lomustine (CCNU) and nimustine (ACNU), which were previously used as standard therapeutic agents against GBM before TMZ, have occasionally been used for the salvage therapy of TMZ‐R‐GBM; however, their efficacy has not yet been thoroughly examined. Therefore, we investigated the antitumor effects of CCNU and ACNU against TMZ‐R‐GBM. As a model of TMZ‐R‐GBM, TMZ resistant clones of human GBM cell lines (U87, U251MG, and U343MG) were established (TMZ‐R‐cells) by the culture of each GBM cells under continuous TMZ treatment, and the antitumor effects of TMZ, CCNU, or ACNU against these cells were analyzed in vitro and in vivo. As a result, although growth arrest and apoptosis were triggered in all TMZ‐R‐cells after the administration of each drug, the antitumor effects of TMZ against TMZ‐R‐cells were significantly reduced compared to those of parental cells, whereas CCNU and ACNU demonstrated efficient antitumor effects on TMZ‐R‐cells as well as parental cells. It was also demonstrated that TMZ resistance of TMZ‐R‐cells was regulated at the initiation of DNA damage response. Furthermore, survival in mice was significantly prolonged by systemic treatment with CCNU or ACNU but not TMZ after implantation of TMZ‐R‐cells. These findings suggest that CCNU or ACNU may serve as a therapeutic agent in salvage treatment against TMZ‐R‐GBM., We investigated the antitumor effects of lomustine (CCNU) and nimustine (ACNU), which were previously used as standard therapeutic agents for glioblastomas (GBM), against the model cells of human GBM cases, which gained acquired temozolomide (TMZ) resistance after continuous treatment by TMZ (TMZ‐R‐cells). We discovered that the antitumor effects of TMZ against TMZ‐R‐cells were significantly reduced compared to those of parental cells, whereas CCNU and ACNU demonstrated efficient antitumor effects on TMZ‐R‐cells as well as parental cells both in vitro and in vivo. In addition, it was also demonstrated that TMZ resistance of TMZ‐R‐cells was regulated at the level of DNA damage response initiation. These findings suggest that CCNU or ACNU may serve as a therapeutic agent in salvage treatment against GBM cases with acquired TMZ resistance.
- Published
- 2021
31. Prognostic value of pre-irradiation FET PET in patients with not completely resectable IDH-wildtype glioma and minimal or absent contrast enhancement
- Author
-
Jan-Michael Werner, Philipp Lohmann, Martin Kocher, Michael Wollring, Karl-Josef Langen, E K Bauer, Gabriele Stoffels, Norbert Galldiks, Gereon R. Fink, Jurij Rosen, Marion Rapp, and Jörg Felsberg
- Subjects
Adult ,Male ,Fluorine Radioisotopes ,Contrast enhancement ,Science ,Article ,Young Adult ,Text mining ,Glioma ,medicine ,Humans ,Aged ,Retrospective Studies ,Univariate analysis ,Multidisciplinary ,Temozolomide ,Receiver operating characteristic ,Brain Neoplasms ,business.industry ,Proportional hazards model ,Wild type ,Middle Aged ,Prognosis ,medicine.disease ,Isocitrate Dehydrogenase ,CNS cancer ,Positron-Emission Tomography ,Tyrosine ,Medicine ,Female ,Cancer in the nervous system ,Nuclear medicine ,business ,ddc:600 ,medicine.drug - Abstract
Scientific reports 11(1), 20828 (2021). doi:10.1038/s41598-021-00193-x, Published by Macmillan Publishers Limited, part of Springer Nature, [London]
- Published
- 2021
32. Clinical features and prognosis of paediatric rhabdomyosarcoma with bone marrow metastasis: a single Centre experiences in China
- Author
-
Xiaoli Ma, Na Xu, Yi Liu, Lejian He, Xue Zhang, Tong Yu, Cheng Huang, Yan Su, Binglin Jian, and Mei Jin
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Bone marrow metastasis ,China ,Adolescent ,Disease ,Pediatrics ,RJ1-570 ,Internal medicine ,Rhabdomyosarcoma ,Medicine ,Humans ,Child ,Retrospective Studies ,Temozolomide ,business.industry ,Research ,Therapeutic effect ,Infant ,Histology ,Retrospective cohort study ,medicine.disease ,Prognosis ,PAX-FOXO1 fusion genes ,Paediatric ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Immunohistochemistry ,Female ,Neoplasm Recurrence, Local ,business ,Bone Marrow Neoplasms ,medicine.drug - Abstract
Background The aim of this study was to summarize the clinical characteristics, therapeutic effects and prognosis of patients with rhabdomyosarcoma (RMS) and bone marrow metastasis, improve the understanding of this disease. Method This was a single-institution retrospective study involving the children with RMS, who presented with bone marrow metastasis at initial presentation to our hospital between 1st, Jan, 2006 and 31st, Dec,2019. Follow-up concluded on 31st, Dec, 2020 and the clinical data were collected and analysed. Result Between 1st Jan 2006 and 31st Dec 2019, 13 eligible patients presented to our hospital, including 10 males and 3 females, these eligible patients accounted for 4.5% of all RMS patients. The median age at onset was 5.6 years (range 1.7-14 years). The patients not only had unfavourable primary sites, but also had multiple metastases. The bone marrow aspirate samples of the patients comprised 8-95% blast-like cells. Nine of 13 patients were misdiagnosed with haematological malignancies or other solid tumours. With respect to histology, four of 13 children were classified as embryonal RMS and nine as alveolar RMS. Eleven patients underwent PAX-FOXO1 fusion testing; eight had the POX- FOXO1 fusion gene. Immunohistochemically(IHC) analysis revealed that the tumour cells were positive for Desmin, Vimentin, Myo-D1 and Myogenin. More importantly, the patients had extremely poor prognoses, the median EFS was 12.0 months (range 3-28.3 months) and the median OS was 27.0 months (range6-46.2 months). Conclusion This study demonstrates that children with RMS and bone marrow metastasis usually exhibit atypical primary sites and multiple metastases, with presentation mimicking haematological malignancies or other solid tumors at initial presentation. Pathology and IHC analysis combined with POX-FOXO1 fusion gene detections can effectively confirm the diagnosis. These patients are more likely to relapse or progress during early treatment and are prone to intracranial metastasis. While multidisciplinary therapy combined with Temozolomide may prevent it, further prospective research is required to evaluate the therapeutic effects.
- Published
- 2021
33. Identification of HOXD10 as a Marker of Poor Prognosis in Glioblastoma Multiforme
- Author
-
Ke Ma, Rongjun Qian, Qi Xie, Xiulei Zhang, Kui Chen, Lingfei Kong, Yanxin Li, and Xianwei Zhang
- Subjects
Oncology ,medicine.medical_specialty ,Temozolomide ,IDH1 ,Competing endogenous RNA ,business.industry ,ceRNA ,medicine.disease ,HOXD10 ,GBM ,OncoTargets and Therapy ,protein-protein interaction ,Internal medicine ,Glioma ,microRNA ,medicine ,Immunohistochemistry ,Pharmacology (medical) ,prognosis ,KEGG ,business ,Original Research ,medicine.drug - Abstract
Yanxin Li,1 Ke Ma,2 Qi Xie,3 Xianwei Zhang,3 Xiulei Zhang,4 Kui Chen,1 Lingfei Kong,3 Rongjun Qian1 1Department of Neurosurgery, Henan Provincial Peopleâs Hospital, Peopleâs Hospital of Zhengzhou University, Peopleâs Hospital of Henan University, Zhengzhou, Henan, 450003, Peopleâs Republic of China; 2Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, Peopleâs Republic of China; 3Department of Pathology, Henan Provincial Peopleâs Hospital, Peopleâs Hospital of Zhengzhou University, Peopleâs Hospital of Henan University, Zhengzhou, Henan, 450003, Peopleâs Republic of China; 4Department of Microbiome Laboratory, Henan Provincial Peopleâs Hospital, Peopleâs Hospital of Zhengzhou University, Peopleâs Hospital of Henan University, Zhengzhou, Henan, 450003, Peopleâs Republic of ChinaCorrespondence: Rongjun QianDepartment of Neurosurgery, Henan Provincial Peopleâs Hospital, Peopleâs Hospital of Zhengzhou University, Peopleâs Hospital of Henan University, No. 7, Wei Wu Road, Zhengzhou, Henan, 450003, Peopleâs Republic of ChinaTel +86 15037138200Email qrjqqx@163.comLingfei KongDepartment of Pathology, Henan Provincial Peopleâs Hospital, Peopleâs Hospital of Zhengzhou University, Peopleâs Hospital of Henan University, No. 7, Wei Wu Road, Zhengzhou, Henan, 450003, Peopleâs Republic of ChinaTel +86 18538298216Email lfkong@zzu.edu.cnPurpose: HOXD10 is a tumor modulator that can either be a tumor-suppressor or a tumor-promoting gene. However, the role of HOXD10 in glioblastoma multiforme (GBM) remains unclear.Methods: Immunohistochemistry (IHC) was applied to detect protein expression of HOXD10 in GBM and normal brain tissue patients. Clinicopathological characteristics with GBM were recorded, and a KaplanâMeier curve was plotted. Additionally, the mRNA expression of HOXD10 and its effect on prognosis were analyzed using the online tool GEPIA and the Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA), and the Gene Expression Omnibus (GEO) databases. Based on the mRNA expression of HOXD10, GBM patients from TCGA database were divided into low- and high-HOXD10 expression groups to analyze the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, and construct a lncRNA-miRNA-mRNA network and a proteinâprotein interaction (PPI) network.Results: The mRNA expression of HOXD10 was up-regulated in GBM according to GEPIA, while the protein expression of HOXD10 in GBM was down-regulated according to IHC analysis of samples from patients collected from our hospital. Correlation analysis showed that HOXD10 expression was significantly related to IDH1 status. Univariate analysis revealed that low HOXD10 expression, complete surgical resection, postoperative radiotherapy, postoperative temozolomide chemotherapy and IDH1 mutation were all beneficial prognostic factors. Further multivariate analysis revealed that only complete surgical resection and postoperative radiotherapy were independent prognostic factors. GO and KEGG enrichment analyses indicated that HOXD10 expression is mainly involved in cytokine-cytokine receptor interactions. In the ceRNA network, 89 nodes, containing 45 mRNAs, 39 miRNAs and five lncRNAs associated with prognosis were involved. The PPI network revealed a tight interaction between HOXD10 and HOXD8, HOXD9, HOXD11, HOXD13 and HOXB3.Conclusion: Based on our experimental data, although HOXD10 expression is low in GBM compared with normal brain tissue, GBM patients with high HOXD10 expression have a worse prognosis. HOXD10 may play different or even opposite roles in different stages of GBM occurrence and development. For patients with GBM, HOXD10 may be a valid predictor of prognosis.Keywords: HOXD10, GBM, prognosis, ceRNA, protein-protein interaction
- Published
- 2021
34. Dose Escalated Radiation Therapy for Glioblastoma Multiforme: An International Systematic Review and Meta-Analysis of 22 Prospective Trials
- Author
-
Raj Singh, Nicholas G. Zaorsky, Joshua D. Palmer, Daniel M. Trifiletti, H.K. Perlow, Ming Wang, Eric J. Lehrer, Silvia Scoccianti, Vinai Gondi, Paul D. Brown, Pierina Navarria, and Joseph Bovi
- Subjects
Adult ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Population ,030218 nuclear medicine & medical imaging ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Epidemiology ,Temozolomide ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Prospective Studies ,Progression-free survival ,Young adult ,Child ,Prospective cohort study ,education ,DNA Modification Methylases ,Aged ,Aged, 80 and over ,education.field_of_study ,Radiation ,Brain Neoplasms ,business.industry ,Tumor Suppressor Proteins ,Radiotherapy Dosage ,Standard of Care ,Middle Aged ,Radiation therapy ,DNA Repair Enzymes ,030220 oncology & carcinogenesis ,Meta-analysis ,Glioblastoma ,business ,medicine.drug - Abstract
Purpose Limited evidence is available on the utility of dose-escalated radiation therapy (DE-RT) with or without temozolomide (TMZ) versus standard-of-care radiation therapy (SoC-RT) for patients with newly diagnosed glioblastoma multiforme. We performed a systematic review/meta-analysis to compare overall survival (OS) and progression-free survival (PFS) between DE-RT and SoC-RT. Methods and Materials We used a Population, Intervention, Control, Outcomes, Study Design/Preferred Reporting Items for Systematic Reviews and Meta-analyses/Meta-analysis of Observational Studies in Epidemiology selection criterion to identify studies. The primary and secondary outcomes were 1-year OS and 1-year PFS, respectively. Outcomes and comparisons were subdivided based on receipt of TMZ and MGMT status. DE-RT was defined based on equivalent dose calculations. Random effects meta-analyses using the Knapp-Hartung correction, arcsine transformation, and restricted maximum likelihood method were conducted. Meta-regression was used to compare therapeutic (eg, DE-RT or TMZ) and pathologic characteristics (eg, MGMT methylation status) using the Wald-type test. Results Across 22 published studies, 2198 patients with glioblastoma multiforme were included; 507 received DE-RT. One-year OS after DE-RT alone was higher than SoC-RT alone (46.3% vs 23.4%; P = .02) as was 1-year PFS (17.9% vs 5.3%; P = .02). No significant difference in 1-year OS (73.2% vs 64.4%; P = .23) or 1-year PFS (44.5% vs 44.3%; P = .33) between DE-RT + TMZ and SoC-RT + TMZ was noted. No difference in 1-year OS was noted between DE-RT + TMZ and SoC-RT + TMZ in either MGMT methylated (83.2% vs 73.2%; P = .23) or MGMT unmethylated (72.6% vs 50.6%; P = .16) patients. Conclusions DE-RT alone resulted in superior PFS and OS versus SoC-RT alone. DE-RT + TMZ did not lead to improved outcomes versus SoC-RT + TMZ. No differential benefit based on MGMT status was found. Future studies are warranted to define which subgroups benefit most from DE-RT.
- Published
- 2021
35. Safety and efficacy of depatuxizumab mafodotin in Japanese patients with malignant glioma: A nonrandomized, phase 1/2 trial
- Author
-
Keisuke Ueki, Masayuki Kanamori, Hao Xiong, Yasuko Nishimura, Motoo Nagane, Yoshihiro Muragaki, Masakazu Yamada, Yoshitaka Narita, Kazuhiko Mishima, Masahide Matsuda, Katsunori Asai, Shota Kasai, Toshihiro Kumabe, Naoki Kagawa, Isao Date, Hiroyuki Kobayashi, Jun-ichiro Kuroda, Christopher Ocampo, and Takaaki Beppu
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,temozolomide ,Antibodies, Monoclonal, Humanized ,Gastroenterology ,Drug Therapy ,Japan ,Clinical Research ,Glioma ,Internal medicine ,medicine ,Clinical endpoint ,Anti–epidermal growth factor receptor therapy ,Humans ,depatuxizumab mafodotin ,Adverse effect ,Aged ,Chemotherapy ,Temozolomide ,business.industry ,Brain Neoplasms ,Incidence (epidemiology) ,Gene Amplification ,General Medicine ,Original Articles ,malignant glioma ,Chemoradiotherapy ,Middle Aged ,medicine.disease ,Survival Analysis ,ErbB Receptors ,Treatment Outcome ,Oncology ,Concomitant ,Original Article ,Female ,Neoplasm Grading ,business ,medicine.drug ,recurrent glioblastoma - Abstract
INTELLANCE‐J was a phase 1/2 study of a potent antibody‐drug conjugate targeting epidermal growth factor receptor (EGFR), depatuxizumab mafodotin (Depatux‐M), as a second‐ or first‐line therapy, alone or combined with chemotherapy or chemoradiotherapy in 53 Japanese patients with World Health Organization (WHO) grade III/IV glioma. In second‐line arms, patients with EGFR‐amplified recurrent WHO grade III/IV glioma received Depatux‐M plus chemotherapy (temozolomide) or Depatux‐M alone regardless of EGFR status. In first‐line arms, patients with newly diagnosed WHO grade III/IV glioma received Depatux‐M plus chemoradiotherapy. The study was halted following lack of survival benefit with first‐line Depatux‐M in the global trial INTELLANCE‐1. The primary endpoint was 6‐month progression‐free survival (PFS) in patients with EGFR‐amplified tumors receiving second‐line Depatux‐M plus chemotherapy. Common nonocular treatment‐emergent adverse events (TEAEs) with both second‐line and first‐line Depatux‐M included lymphopenia (42%, 33%, respectively), thrombocytopenia (39%, 47%), alanine aminotransferase increase (29%, 47%), and aspartate aminotransferase increase (24%, 60%); incidence of grade ≥3 TEAEs was 66% and 53%, respectively. Ocular side effects (OSEs) occurred in 93% of patients receiving second‐line Depatux‐M plus chemotherapy and all patients receiving second‐line Depatux‐M alone or first‐line Depatux‐M plus chemoradiotherapy. Most OSEs were manageable with dose modifications and concomitant medications. The 6‐month PFS estimate was 25.6% (95% confidence interval [CI] 11.4‒42.6), and median PFS was 2.1 months (95% CI 1.9‒3.9) with second‐line Depatux‐M plus chemotherapy in the EGFR‐amplified subgroup. This study showed acceptable safety profile of Depatux‐M alone or plus chemotherapy/chemoradiotherapy in Japanese patients with WHO grade III/IV glioma. The study was registered at ClinicalTrials.gov (NCT02590263)., INTELLANCE‐J was a phase 1/2 study of a potent antibody‐drug conjugate targeting epidermal growth factor receptor (EGFR), depatuxizumab mafodotin, as a second‐ or first‐line therapy, alone or combined with chemotherapy or chemoradiotherapy in Japanese patients with World Health Organization grade III/IV glioma. The results of this trial demonstrate an acceptable safety profile of depatuxizumab mafodotin, with ocular side effects being the most common adverse events that were mostly reversible. Second‐line depatuxizumab mafodotin in combination with temozolomide resulted in a 6‐month progression‐free survival estimate of 25.6% (95% confidence interval 11.4‒42.6) in patients with EGFR‐amplified tumors and showed encouraging antitumor activity in this subgroup of patients (NCT02590263).
- Published
- 2021
36. Accurately Controlled Delivery of Temozolomide by Biocompatible UiO-66-NH2 Through Ultrasound to Enhance the Antitumor Efficacy and Attenuate the Toxicity for Treatment of Malignant Glioma
- Author
-
Jiahao Jiang, Jun Qian, Wan Zhiping, Gu Jinmao, Jiaqi Wang, Chunlin Li, Yinwen Li, Zhu Junle, Chun Luo, and Chen Huairui
- Subjects
Medicine (General) ,Biocompatibility ,Biophysics ,Pharmaceutical Science ,Bioengineering ,Biomaterials ,ultrasound glioma ,R5-920 ,International Journal of Nanomedicine ,In vivo ,Glioma ,Drug Discovery ,medicine ,tmz ,Temozolomide ,Chemistry ,business.industry ,Organic Chemistry ,Therapeutic effect ,Ultrasound ,General Medicine ,medicine.disease ,Bone marrow suppression ,Toxicity ,Cancer research ,business ,mof ,medicine.drug - Abstract
Zhiping Wan,1,* Chunlin Li,2,* Jinmao Gu,1,* Jun Qian,1 Junle Zhu,1 Jiaqi Wang,2 Yinwen Li,2 Jiahao Jiang,1 Huairui Chen,1 Chun Luo1 1Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200092, Peopleâs Republic of China; 2Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, Peopleâs Republic of China*These authors contributed equally to this workCorrespondence: Chun Luo; Huairui Chen Email boyluochun@126.com; chen13761626536@163.comBackground: Glioma is the most common and malignant primary brain tumour in adults and has a dismal prognosis. Temozolomide (TMZ) is the only clinical first-line chemotherapy drug for malignant glioma up to present. Due to poor aqueous solubility and toxic effects, TMZ is still inefficient and limited for clinical glioma treatment.Methods: UiO-66-NH2 nanoparticle is a zirconium-based framework, constructed by Zr and 2-amino-1,4-benzenedicarboxylic acid (BDC-NH2) with octahedral microporous structure, which can be decomposed by the body into an ionic form to discharge. We prepared the nanoscale metal-organic framework (MOF) of UiO-66-NH2 to load TMZ for therapy of malignant glioma, TMZ is released from UiO-66-NH2 through a porous structure. The ultrasound accelerates its porous percolation and promotes the rapid dissolution of TMZ through low-frequency oscillations and cavitation effect. The biological safety and antitumor efficacy were evaluated both in vitro and in vivo.Results: The prepared TMZ@MOF exhibited excellent biocompatibility and biosafety due to minimal drug leakage without ultrasound intervention. We further used the flank model of glioblastoma to verify the in vivo therapeutic effect. TMZ@UiO-66-NH2 nanocomposites could be well delivered to the tumour tissue, which led to local enrichment of the TMZ concentration. Furthermore, TMZ@UiO-66-NH2 nanocomposites under ultrasound demonstrated much more efficient inhibition for tumor growth than TMZ@UiO-66-NH2 nanocomposites and TMZ alone. Meanwhile, the bone marrow suppression side effects of TMZ were significantly reduced by TMZ@UiO-66-NH2 nanocomposites.Conclusion: In this work, TMZ@UiO-66-NH2 nanocomposites with ultrasound mediation could effectively improve the killing effect of malignant glioma and decrease TMZ-induced toxicity in normal tissues, demonstrating great potential for the delivery of TMZ in the clinical treatment of malignant gliomas.Keywords: MOF, TMZ, ultrasound glioma
- Published
- 2021
37. Glioma-Targeted Therapeutics: Computer-Aided Drug Design Prospective
- Author
-
Mahmoud E. S. Soliman, Clement Agoni, Preantha Poonan, and Mahmoud A. A. Ibrahim
- Subjects
Oncology ,Drug ,Chemotherapy ,medicine.medical_specialty ,Temozolomide ,Drug discovery ,business.industry ,medicine.medical_treatment ,media_common.quotation_subject ,Organic Chemistry ,Cancer ,Bioengineering ,Disease ,medicine.disease ,Biochemistry ,Analytical Chemistry ,Glioma ,Internal medicine ,medicine ,business ,Relevant information ,medicine.drug ,media_common - Abstract
Amongst the several types of brain cancers known to humankind, glioma is one of the most severe and life-threatening types of cancer, comprising 40% of all primary brain tumors. Recent reports have shown the incident rate of gliomas to be 6 per 100,000 individuals per year globally. Despite the various therapeutics used in the treatment of glioma, patient survival rate remains at a median of 15 months after undergoing first-line treatment including surgery, radiation, and chemotherapy with Temozolomide. As such, the discovery of newer and more effective therapeutic agents is imperative for patient survival rate. The advent of computer-aided drug design in the development of drug discovery has emerged as a powerful means to ascertain potential hit compounds with distinctively high therapeutic effectiveness against glioma. This review encompasses the recent advances of bio-computational in-silico modeling that have elicited the discovery of small molecule inhibitors and/or drugs against various therapeutic targets in glioma. The relevant information provided in this report will assist researchers, especially in the drug design domains, to develop more effective therapeutics against this global disease.
- Published
- 2021
38. Prognostic Impact of the Combination of MGMT Methylation and TERT Promoter Mutation in Glioblastoma
- Author
-
Surasak Sangkhathat, Pimwara Tanvejsilp, Kanet Kanjanapradit, and Thara Tunthanathip
- Subjects
Oncology ,medicine.medical_specialty ,Methyltransferase ,medicine.medical_treatment ,Neurosciences. Biological psychiatry. Neuropsychiatry ,survival ,o6-methylguanine-dna methyltransferase ,Internal medicine ,medicine ,Telomerase reverse transcriptase ,neoplasms ,Survival analysis ,Temozolomide ,business.industry ,Proportional hazards model ,General Neuroscience ,telomerase reverse transcriptase ,O-6-methylguanine-DNA methyltransferase ,Nomogram ,digestive system diseases ,Radiation therapy ,primary glioblastoma ,prognosis ,Neurology (clinical) ,business ,RC321-571 ,medicine.drug - Abstract
Background The concept of combinational analysis between the methylation of O6-methylguanine-DNA methyltransferase (MGMT) and telomerase reverse transcriptase promoter (pTERT) mutation in glioblastoma (GBM) has been reported. The main study objective was to determine the prognosis of patients with GBM based on MGMT/pTERT classification, while the secondary objective was to estimate the temozolomide effect on the survival time of GBM with MGMT/pTERT classification. Methods A total of 50 GBM specimens were collected after tumor resection and were selected for investigating MGMT methylation and pTERT mutation. Clinical imaging and pathological characteristics were retrospectively analyzed. Patients with MGMT/pTERT classification were analyzed using survival analysis to develop the nomogram for forecasting and individual prognosis. Results All patients underwent resection (total resection: 28%, partial resection: 64%, biopsy: 8%). Thirty-two percent of all cases received adjuvant temozolomide with radiotherapy. Sixty-four percent of the case was found methylated MGMT, and 56% of the present cohort found pTERT mutation. Following combinational analysis of biomarkers, results showed that the GBMs with methylated MGMT and wild-type pTERT had a superior prognosis compared with other subtypes. Using Cox regression analysis with multivariable analysis, the extent of resection, postoperative chemoradiotherapy, MGMT/pTERT classification were associated with a favorable prognosis. Hence, a web-based nomogram was developed for deploying individual prognostication. Conclusions The interaction of MGMT methylation and pTERT mutation was confirmed for predicting prognosis. The results from the present study could help physicians create treatment strategies for GBM patients in real-world situations.
- Published
- 2021
39. Quantification of SPECT with 99mTc-technetryl and of contrast-enhanced MRI scans in survival prognosis of patients with glial brain tumors after combined chemo – radiation treatment
- Subjects
Chemotherapy ,Temozolomide ,Radiological and Ultrasound Technology ,medicine.diagnostic_test ,business.industry ,Adjuvant chemotherapy ,medicine.medical_treatment ,Magnetic resonance imaging ,Glial tumor ,Concomitant ,medicine ,Radiology, Nuclear Medicine and imaging ,Nuclear medicine ,business ,Emission computed tomography ,Chemoradiotherapy ,medicine.drug - Abstract
Purpose of study. The possibility of simultaneous and combined use of magnetic resonance imaging (MRI) with paramagnetic contrast enhancement (CE-MRI) and single-photon emission computed tomography (SPECT) with 99mTc-MIBI in assessing the prognosis in patients with glial brain tumors after complex chemo – and radiotherapy treatment was studied.Material and methods. Contrast-enhanced MRI of the brain and SPECT with 99mTc-Technetril were performed in 19 patients who were observed after complex chemoradiotherapy of brain tumors (gliomas of grade 3 and 4). The treatment included remote gamma irradiation and chemotherapy with temozolomide. The radiation dose was 60 Gy (30 fractions of 2 Gy × 1 once a day). The drug Temodal was prescribed in a single dose of 75 mg/sq. m. (120–140 mg) an hour before gamma therapy for up to 40 days (in a total dose of 5000–5900 mg) and in the post – radiation period according to the scheme – 6 courses of the drug for 5 days every 23 days in a single dose of 200 mg/sq.m. (280–400 mg). The total dose of Temodal for 1 course was 1400–2000 mg. In everybody, after the study, the time period of subsequent survival of patients was registered from to the primary health care data.Results. The survival time was over 20 months in case of patients after surgical removal of glial tumor and with subsequent adjuvant chemotherapy if simultaneous low values of “Tumor-to Normal Tissue” index for both 99mTc-Technetryl SPECT and CE-MRI were met, i.e. if < 1,15 for 99mTc-Technetryl SPECT and < 1,19 for CE-MRI. If index “Tumor-to Normal Tissue” was over > 1,25 99mTc-Technetryl SPECT and > 1,35 for CE-MRI then the survival time was below one year. If the “Tumor-to Normal Tissue”ratio on 99mTc-Technetryl SPECT scan was in benefit ranges whereas the on CE-MRI kept itself in high pathologic ranges the survival time was also over 19 months.Conclusion. Thus, the concomitant quantification and fusion of uptake of 99mTc-Technetryl and paramagnetic contrast agents on SPECT and MRI scans provides prognostic data on survival of patients and is worth routine use for therapy control.
- Published
- 2021
40. Adenosine A2A Receptor Activation Enhances Blood–Tumor Barrier Permeability in a Rodent Glioma Model
- Author
-
Jessica Bills, Meili Zhang, Dionne Davis, Monica Manglani, Kunio Nagashima, Stuart Walbridge, Brandon Foster, Dorian B. McGavern, Priya Shankarappa, Amélie Vézina, DreeAnna Morris, Matthew McCord, Hua Song, Jessica Kindrick, William D. Figg, Wei Zhang, Leslie L. Muldoon, Sadhana Jackson, Mark R. Gilbert, and Cody J. Peer
- Subjects
Cancer Research ,Receptor, Adenosine A2A ,Adenosine A2A receptor ,Mice, SCID ,Transfection ,Article ,Mice ,Rats, Nude ,Downregulation and upregulation ,Glioma ,Animals ,Humans ,Medicine ,Molecular Biology ,Temozolomide ,Brain Neoplasms ,business.industry ,medicine.disease ,Survival Analysis ,Rats ,Regadenoson ,Endothelial stem cell ,Disease Models, Animal ,Oncology ,Blood-Brain Barrier ,Paracellular transport ,Cancer research ,Female ,Stem cell ,business ,medicine.drug - Abstract
The blood–tumor barrier (BTB) limits the entry of effective chemotherapeutic agents into the brain for treatment of malignant tumors like glioblastoma. Poor drug entry across the BTB allows infiltrative glioma stem cells to evade therapy and develop treatment resistance. Regadenoson, an FDA-approved adenosine A2A receptor (A2AR) agonist, has been shown to increase drug delivery across the blood–brain barrier in non–tumor-bearing rodents without a defined mechanism of enhancing BTB permeability. Here, we characterize the time-dependent impact of regadenoson on brain endothelial cell interactions and paracellular transport, using mouse and rat brain endothelial cells and tumor models. In vitro, A2AR activation leads to disorganization of cytoskeletal actin filaments by 30 minutes, downregulation of junctional protein expression by 4 hours, and reestablishment of endothelial cell integrity by 8 hours. In rats bearing intracranial gliomas, regadenoson treatment results in increase of intratumoral temozolomide concentrations, yet no increased survival noted with combined temozolomide therapy. These findings demonstrate regadenoson's ability to induce brain endothelial structural changes among glioma to increase BTB permeability. The use of vasoactive mediators, like regadenoson, which transiently influences paracellular transport, should further be explored to evaluate their potential to enhance central nervous system treatment delivery to aggressive brain tumors. Implications: This study provides insight on the use of a vasoactive agent to increase exposure of the BTB to chemotherapy with intention to improve glioma treatment efficacy.
- Published
- 2021
41. Celecoxib reverses the glioblastoma chemo-resistance to temozolomide through mitochondrial metabolism
- Author
-
Jing Zhao, Wei Zhou, Chen Gong, Guoqing Jin, Hong He, Zhenbo Fan, Huihua Xiong, and Delong Yin
- Subjects
Aging ,mitochondrial metabolism ,Combination therapy ,Cell Survival ,Angiogenesis ,medicine.medical_treatment ,Respiratory chain ,temozolomide ,chemo-resistance ,Cell Line, Tumor ,medicine ,Humans ,Phosphorylation ,Chemotherapy ,Temozolomide ,Cell growth ,business.industry ,glioblastoma ,Cell Biology ,Mitochondria ,cyclooxygenase-2 ,Celecoxib ,Drug Resistance, Neoplasm ,Apoptosis ,Cancer research ,Drug Therapy, Combination ,business ,Oxidation-Reduction ,Research Paper ,medicine.drug - Abstract
Temozolomide (TMZ) is used for the treatment of high-grade gliomas. Acquired chemoresistance is a serious limitation to the therapy with more than 90% of recurrent gliomas showing little response to a second line of chemotherapy. Therefore, it is necessary to explore an alternative strategy to enhance the sensitivity of glioblastoma (GBM) to TMZ in neuro-oncology. Celecoxib is well known and widely used in anti-inflammatory and analgesic. Cyclooxygenase-2 (COX-2) expression has been linked to the prognosis, angiogenesis, and radiation sensitivity of many malignancies such as primitive neuroectodermal tumor and advanced melanoma. The objective of this study was to explore the chemotherapy-sensitizing effect of celecoxib on TMZ in GBM cells and its potential mechanisms. From the study, we found that the combination therapy (TMZ 250uM+celecoxib 30uM) showed excellent inhibitory effect to the GBM, the LN229 and LN18, which were the TMZ resistant GBM cell lines. Our data suggest that the combination therapy may inhibits cell proliferation, increases apoptosis, and increases the autophagy on LN229 and LN18. The potential molecular mechanisms were related to mitochondrial metabolism and respiratory chain inhibition.
- Published
- 2021
42. Calcyphosine promotes the proliferation of glioma cells and serves as a potential therapeutic target
- Author
-
Zhi-Cheng He, Xiao-Qing Xie, Xiu-Wu Bian, Qing Liu, Jiao Wang, Zheng Zhu, Juan Tan, Yue-Liang Yao, Tao Luo, Ze-Xuan Yan, Wen-Juan Fu, and Yan-Xia Wang
- Subjects
Adult ,Male ,Chemosensitizer ,Apoptosis ,PLK1 ,Pathology and Forensic Medicine ,Mice ,chemistry.chemical_compound ,Glioma ,medicine ,Animals ,Humans ,Aged ,Cell Proliferation ,Gene knockdown ,Temozolomide ,Brain Neoplasms ,business.industry ,Pteridines ,Calcium-Binding Proteins ,Cell Cycle ,Volasertib ,Middle Aged ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,chemistry ,Cancer research ,Female ,business ,medicine.drug - Abstract
Calcyphosine (CAPS) was initially identified from the canine thyroid. It also exists in many types of tumor, but its expression and function in glioma remain unknown. Here we explored the clinical significance and the functional mechanisms of CAPS in glioma. We found that CAPS was highly expressed in glioma and high expression of CAPS was correlated with poor survival, in glioma patients and public databases. Cox regression analysis showed that CAPS was an independent prognostic factor for glioma patients. Knockdown of CAPS suppressed the proliferation, whereas overexpression of CAPS promoted the proliferation of glioma both in vitro and in vivo. CAPS regulated the G2/M phase transition of the cell cycle, but had no obvious effect on apoptosis. CAPS affected PLK1 phosphorylation through interaction with MYPT1. CAPS knockdown decreased p-MYPT1 at S507 and p-PLK1 at S210. Expression of MYPT1 S507 phosphomimic rescued PLK1 phosphorylation and the phenotype caused by CAPS knockdown. The PLK1 inhibitor volasertib enhanced the therapeutic effect of temozolomide in glioma. Our data suggest that CAPS promotes the proliferation of glioma by regulating the cell cycle and the PLK1 inhibitor volasertib might be a chemosensitizer of glioma. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
- Published
- 2021
43. Cushing’s syndrome due to adrenocorticotropic hormone-secreting metastatic neuroendocrine tumor of unknown primary origin: a case report and literature review
- Author
-
Hayri Bostan, Ayse Kevser Erdol, Muhammed Erkam Sencar, Murat Calapkulu, Serap Duru, Pinar Akhanli, Muhammed Kizilgul, Tugba Taskin Turkmenoglu, Erman Cakal, Mustafa Ozbek, Hakan Duger, and Bekir Ucan
- Subjects
Adult ,Male ,Teaching Case Presentations ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Proliferation index ,Ectopic Cushing’s syndrome ,Endocrinology, Diabetes and Metabolism ,Carcinoid Tumor ,Adrenocorticotropic hormone ,Capecitabine ,Lesion ,Adrenocorticotropic Hormone ,Positron Emission Tomography Computed Tomography ,Temozolomide ,medicine ,Humans ,Ga-68 labeled somatostatin receptor PET/CT ,Cushing Syndrome ,business.industry ,Unknown primary origin ,COVID-19 ,General Medicine ,Inferior petrosal sinus sampling ,Neuroendocrine neoplasm ,ACTH Syndrome, Ectopic ,Neuroendocrine Tumors ,Hypercortisolemia ,Somatostatin ,Atypical pulmonary carcinoid ,Dexamethasone suppression test ,Neoplasms, Unknown Primary ,medicine.symptom ,business ,medicine.drug - Abstract
Background In this article, we present a case of neuroendocrine neoplasm of unknown primary origin (UPO NEN), which is a rare cause of ectopic Cushing’s syndrome (ECS) presenting numerous challenges, together with a literature review. Case report A 43-year-old male patient presented with clinical features consistent with Cushing’s syndrome (CS) and adrenocorticotropic hormone (ACTH)-dependent hypercortisolemia. Despite a suspicious lesion on pituitary MRI, the high-dose dexamethasone suppression test and bilateral inferior petrosal sinus sampling results were not compatible with Cushing’s disease. Bilateral non-homogeneous opacities were observed in the thorax CT of the patient, who also had a history of COVID-19 infection, but no tumoral lesion was detected. When 68Ga-SSTR PET/CT and 18FDG-PET/CT were performed, multiple metastatic foci were detected in mediastinal and hilar lymph nodes and the axial skeleton. Paratracheal-subcarinal lymph nodes were excised mediastinoscopically, and the diagnosis of NEN was made. Histopathological findings indicated that the possible origin was an atypical pulmonary carcinoid with a low Ki-67 labeling index. After controlling hypercortisolemia, a regimen of somatostatin analogs and capecitabine plus temozolomide was decided upon as treatment by a multidisciplinary council. Conclusion This is a challenging case of UPO NEN presenting with ECS and confounding factors, such as previous infection and incidental lesions, during the diagnosis process. The case in question highlighted the fact that atypical pulmonary carcinoid with a low proliferation index may cause visible metastases even when radiologically undetectable.
- Published
- 2021
44. Prediction of Glioma Stemlike Cell Infiltration in the Non–Contrast-Enhancing Area by Quantitative Measurement of Lactate on Magnetic Resonance Spectroscopy in Glioblastoma
- Author
-
Shirabe Matsumoto, Seiji Shigekawa, Hideaki Watanabe, Saya Ozaki, Yoshihiro Ohtsuka, Hajime Yano, Akihiro Inoue, Takanori Ohnishi, Riko Kitazawa, Yawara Nakamura, Junya Tanaka, Takeharu Kunieda, Yonehiro Kanemura, Masahiro Nishikawa, Daisuke Yamashita, and Satoshi Suehiro
- Subjects
Adult ,Male ,Magnetic Resonance Spectroscopy ,Lactate dehydrogenase A ,Cell ,Carbohydrate metabolism ,Creatine ,Neurosurgical Procedures ,Young Adult ,chemistry.chemical_compound ,Methionine ,Glioma ,Temozolomide ,Humans ,Medicine ,Pyruvate Dehydrogenase (Lipoamide) ,Lactic Acid ,RNA, Messenger ,education ,Antineoplastic Agents, Alkylating ,Aged ,Aged, 80 and over ,education.field_of_study ,biology ,Brain Neoplasms ,business.industry ,CD44 ,Chemoradiotherapy, Adjuvant ,Middle Aged ,Pyruvate dehydrogenase complex ,medicine.disease ,Magnetic Resonance Imaging ,Mitochondria ,nervous system diseases ,medicine.anatomical_structure ,chemistry ,Anaerobic glycolysis ,Positron-Emission Tomography ,Neoplastic Stem Cells ,Cancer research ,biology.protein ,Female ,Surgery ,Neurology (clinical) ,Lactate Dehydrogenase 5 ,Radiopharmaceuticals ,Energy Metabolism ,Glioblastoma ,business - Abstract
Background We previously reported that glioma stemlike cells (GSCs) exist in the area of the tumor periphery showing no gadolinium enhancement on magnetic resonance imaging. In the present work, we analyzed glucose metabolism to investigate whether lactate could be predictive of tumor invasiveness and of use in detection of the tumor invasion area in glioblastoma multiforme (GBM). Methods The expression of lactate dehydrogenase A (LDH-A) and pyruvate dehydrogenase (PDH) was investigated in 20 patients. In GSC lines, LDH-A and PDH expression also was examined in parallel to assessments of mitochondrial respiration. We then investigated the relationship between lactate/creatine ratios in the tumor periphery measured by magnetic resonance spectroscopy, using learning-compression-model algorithms and phenotypes of GBMs. Results In 20 GBMs, high-invasive GBM expressed LDH-A at significantly higher expression than did low-invasive GBM, whereas low-invasive GBM showed significantly higher expression of PDH than did high-invasive GBM. The highly invasive GSC line showed higher expression of LDH-A and lower expression of PDH compared with low-invasive GSC lines. The highly invasive GSC line also showed the lowest consumption of oxygen and the lowest production of adenosine triphosphate. Lactate levels, as measured by magnetic resonance spectroscopy, showed a significant positive correlation with LDH-A transcript levels, permitting classification of the GBMs into high-invasive and low-invasive phenotypes based on a cutoff value of 0.66 in the lactate/creatine ratio. Conclusions In the tumor periphery area of the highly invasive GBM, aerobic glycolysis was the predominant pathway for glucose metabolism, resulting in the accumulation of lactate. The level of lactate may facilitate prediction of the tumor-infiltrating area on GBM.
- Published
- 2021
45. Adding high-dose celecoxib to increase effectiveness of standard glioblastoma chemoirradiation
- Author
-
Richard E. Kast
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Short paper ,Pharmaceutical Science ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Generic drug ,Temozolomide ,medicine ,Humans ,neoplasms ,Pharmacology ,biology ,business.industry ,medicine.disease ,Clinical trial ,030104 developmental biology ,Celecoxib ,Cyclooxygenase 2 ,030220 oncology & carcinogenesis ,Pharmacodynamics ,biology.protein ,Cyclooxygenase ,Glioblastoma ,business ,medicine.drug - Abstract
Over one hundred clinical trials since 2005 have failed to significantly improve the prognosis of glioblastoma. Since 2005, the standard of care has been maximal resection followed by 60Gy irradiation over six weeks with daily temozolomide. With this, a median survival of 2 years can be expected. This short paper reviewed how the pharmacodynamic attributes of an EMA/FDA approved, cheap, generic drug to treat pain, celecoxib, intersect with pathophysiological elements driving glioblastoma growth, such that growth drive inhibition can be expected from celecoxib. The two main attributes of celecoxib are carbonic anhydrase inhibition and cyclooxygenase-2 inhibition. Both attributes individually have been in active study as adjuncts during current cancer treatment, including that of glioblastoma. That research is briefly reviewed here. This paper concludes from the collected data, that starting celecoxib, 600 to 800mg twice daily before surgery and continuing it through the chemoirradiation phase of treatment would be a low-risk intervention with sound rationale.
- Published
- 2021
46. Pulsed and Discontinuous Electromagnetic Field Exposure Decreases Temozolomide Resistance in Glioblastoma by Modulating the Expression of O6-Methylguanine-DNA Methyltransferase, Cyclin-D1, and p53
- Author
-
Lily Mohammadipoor-ghasemabad, Abdolreza Babaee, Parisa Vosough, Mohsen Basiri, Samereh Dehghani-Soltani, Meysam Ahmadi-Zeidabadi, and Seyed Hassan Eftekhar-Vaghefi
- Subjects
0301 basic medicine ,Pharmacology ,Cancer Research ,Chemotherapy ,Temozolomide ,business.industry ,medicine.medical_treatment ,Brain tumor ,General Medicine ,medicine.disease ,DNA methyltransferase ,Radiation therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cyclin D1 ,Oncology ,030220 oncology & carcinogenesis ,Concomitant ,medicine ,Cancer research ,Radiology, Nuclear Medicine and imaging ,business ,medicine.drug ,Glioblastoma - Abstract
Background: Glioblastoma is a malignant and very aggressive brain tumor with a poor prognosis. Despite having chemotherapy concomitant with surgery and/or radiation therapy, the median survival of ...
- Published
- 2021
47. Imipramine impedes glioma progression by inhibiting YAP as a Hippo pathway independent manner and synergizes with temozolomide
- Author
-
Di Wu, Rutong Yu, Ji Qi, Qingming Meng, Yu Zhang, Kai Wang, Qiang Wang, Xu Wang, Xiang Wang, Xiuping Zhou, Er Nie, Yan Wang, and Ding Zhou
- Subjects
Imipramine ,Combination therapy ,medicine.drug_class ,proliferation ,Tricyclic antidepressant ,Antineoplastic Agents ,Cell Cycle Proteins ,temozolomide ,Protein Serine-Threonine Kinases ,Prostate cancer ,Mice ,Glioma ,glioma ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cell Proliferation ,Hippo signaling pathway ,Temozolomide ,Oncogene ,Dose-Response Relationship, Drug ,business.industry ,Intracellular Signaling Peptides and Proteins ,Drug Synergism ,Cell Biology ,Original Articles ,medicine.disease ,Prognosis ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,Treatment Outcome ,Cancer research ,Molecular Medicine ,Original Article ,YAP ,business ,medicine.drug ,Signal Transduction ,Transcription Factors - Abstract
Patients with malignant glioma often suffered from depression, which leads to an increased risk of detrimental outcomes. Imipramine, an FDA‐approved tricyclic antidepressant, has been commonly used to relieve depressive symptoms in the clinic. Recently, imipramine has been reported to participate in the suppression of tumour progression in several human cancers, including prostate cancer, colon cancer and lymphomas. However, the effect of imipramine on malignant glioma is largely unclear. Here, we show that imipramine significantly retarded proliferation of immortalized and primary glioma cells. Mechanistically, imipramine suppressed tumour proliferation by inhibiting yes‐associated protein (YAP), a recognized oncogene in glioma, independent of Hippo pathway. In addition to inhibiting YAP transcription, imipramine also promoted the subcellular translocation of YAP from nucleus into cytoplasm. Consistently, imipramine administration significantly reduced orthotopic tumour progression and prolonged survival of tumour‐bearing mice. Moreover, exogenous overexpression of YAP partially restored the inhibitory effect of imipramine on glioma progression. Most importantly, compared with imipramine or temozolomide (TMZ) monotherapy, combination therapy with imipramine and TMZ exhibited enhanced inhibitory effect on glioma growth both in vitro and in vivo, suggesting the synergism of both agents. In conclusion, we found that tricyclic antidepressant imipramine impedes glioma progression by inhibiting YAP. In addition, combination therapy with imipramine and TMZ may potentially serve as promising anti‐glioma regimens, thus predicting a broad prospect of clinical application.
- Published
- 2021
48. Efficacy and safety of nivolumab in Japanese patients with first recurrence of glioblastoma: an open-label, non-comparative study
- Author
-
Mitsutoshi Nakada, Naoki Kagawa, Manabu Natsumeda, Shota Tanaka, Yukihiko Sonoda, Yasuo Iwadate, Tomokazu Aoki, Nobuhiro Hata, Hironobu Minami, Yuki Hirata, Shigeru Yamaguchi, Yoshiki Arakawa, Satoshi Suehiro, Kazuhiko Sugiyama, Toshihiko Wakabayashi, Yoichi Nakazato, Shunsuke Hagihara, Jun-ichiro Kuroda, Yoshitaka Narita, Yoshihiro Muragaki, Motoo Nagane, Ryo Nishikawa, and Eiichi Ishikawa
- Subjects
Oncology ,medicine.medical_specialty ,Gliosarcoma ,Bevacizumab ,Phases of clinical research ,Japan ,Internal medicine ,Glioma ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Programmed cell death ,Temozolomide ,business.industry ,Bayesian approach ,Bayes Theorem ,Hematology ,General Medicine ,medicine.disease ,Clinical Trial ,Confidence interval ,Phase II ,Nivolumab ,Surgery ,Original Article ,Neoplasm Recurrence, Local ,business ,Glioblastoma ,medicine.drug - Abstract
Background An open-label, non-comparative study assessed the efficacy and safety of nivolumab in Japanese patients with first recurrence glioblastoma. Methods Patients with first recurrence of histologically confirmed World Health Organization Grade IV glioma, after treatment with temozolomide and radiotherapy, received nivolumab 3 mg/kg every 2 weeks until confirmed disease progression (Response Assessment in Neuro-Oncology criteria) or toxicity. Primary endpoint was 1-year overall survival rate assessed by Bayesian approach. The prespecified efficacy criterion was that the Bayesian posterior probability threshold for exceeding the 1-year overall survival of bevacizumab (34.5%) from the Japanese phase 2 study (JO22506) would be 93%. Results Of the 50 enrolled patients, 44 (88.0%) had recurrent malignant glioma (glioblastoma, gliosarcoma), and of these, 26 (59.1%) had at least one measurable lesion at baseline. The Bayesian posterior mean 1-year overall survival (90% Bayesian credible intervals) with nivolumab was 54.4% (42.27–66.21), and the Bayesian posterior probability of exceeding the threshold of the 1-year overall survival rate of bevacizumab (34.5%) was 99.7%. Median (90% confidence interval) overall and progression-free survival was 13.1 (10.4–17.7) and 1.5 (1.4–1.5) months, respectively. One partial response was observed (objective response rate 1/26 evaluable patients [3.8%]). Treatment-related adverse event rates were 14.0% for Grade 3–4 and 2.0% for Grade 5; most adverse events resolved and were manageable. Conclusions The 1-year overall survival with nivolumab monotherapy in Japanese patients with glioblastoma met the prespecified efficacy criterion. The safety profile of nivolumab was consistent with that observed in other tumor types. Clinical Trial Registration JapicCTI-152967.
- Published
- 2021
49. Sex-based survival differences in IDH-wildtype glioblastoma: Results from a retrospective cohort study
- Author
-
Tom Boterberg, J.P. Kalala, D. Van Roost, C. Van den Broecke, Harry Pinson, and Giorgio Hallaert
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Neurology ,Physiology (medical) ,Internal medicine ,medicine ,Humans ,DNA Modification Methylases ,Survival analysis ,Retrospective Studies ,Temozolomide ,Brain Neoplasms ,Proportional hazards model ,business.industry ,Retrospective cohort study ,General Medicine ,DNA Methylation ,Prognosis ,Isocitrate Dehydrogenase ,DNA Repair Enzymes ,Isocitrate dehydrogenase ,Cohort ,Female ,Surgery ,Neurology (clinical) ,Glioblastoma ,business ,Chemoradiotherapy ,medicine.drug - Abstract
A female survival benefit has been described for glioblastoma patients. Recent studies report that the effect of 06-methylguanine-DNA-methyltransferase gene promoter (MGMTp) methylation is only present in female patients. We retrospectively studied sex-based survival, including MGMTp-methylation, in a cohort of 159 uniformly treated isocitrate dehydrogenase wildtype (IDHwt) patients. All patients were treated with temozolomide-based chemoradiotherapy after surgery. Kaplan-Meier survival curves and Cox regression models were used to evaluate overall survival. The study included 59 female (37.1%) and 100 male patients (62.9%). There were no statistically significant differences between sexes concerning demographic, surgical or radiological characteristics. Female patients harbored MGMTp-methylated tumors in 45.8% of cases and males in 33% (P = 0.129). Median overall survival was 13.4 months for men and women alike. After adjustment of survival for age, Karnofsky Performance Score, extent of resection and MGMTp-methylation, sex did not have a significant survival impact. However, MGMTp-methylation proved to be an independent beneficial prognosticator for both sexes, contradicting earlier reports. Several sex-based molecular subtypes of glioblastoma with different response to current treatment may exist explaining conflicting survival results in different patient cohorts. Further research on sex-based differences in IDHwt glioblastoma patients is needed.
- Published
- 2021
50. Exosomal circ-HIPK3 Facilitates Tumor Progression and Temozolomide Resistance by Regulating miR-421/ZIC5 Axis in Glioma
- Author
-
Chengchen Han, Jianning Zhang, Shuwei Wang, and Hongwei Wang
- Subjects
0301 basic medicine ,Pharmacology ,Cancer Research ,Temozolomide ,business.industry ,General Medicine ,medicine.disease ,nervous system diseases ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Tumor progression ,Circular RNA ,030220 oncology & carcinogenesis ,Glioma ,Cancer research ,Medicine ,Radiology, Nuclear Medicine and imaging ,business ,neoplasms ,Clinical treatment ,medicine.drug - Abstract
Background: The resistance of glioma patients to temozolomide (TMZ) treatment is a limiting factor in clinical treatment. Circular RNA HIPK3 (circ-HIPK3) was found to be highly expressed in glioma,...
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.