cytotoxic treatment
Recently Published Documents


TOTAL DOCUMENTS

215
(FIVE YEARS 51)

H-INDEX

33
(FIVE YEARS 3)

2022 ◽  
Vol 12 ◽  
Author(s):  
Jinguo Zhang ◽  
Shuaikang Pan ◽  
Chen Jian ◽  
Li Hao ◽  
Jie Dong ◽  
...  

Breast cancer (BC) is the most common malignancy among females. Chemotherapy drugs remain the cornerstone of treatment of BC and undergo significant shifts over the past 100 years. The advent of immunotherapy presents promising opportunities and constitutes a significant complementary to existing therapeutic strategies for BC. Chemotherapy as a cytotoxic treatment that targets proliferation malignant cells has recently been shown as an effective immune-stimulus in multiple ways. Chemotherapeutic drugs can cause the release of damage-associated molecular patterns (DAMPs) from dying tumor cells, which result in long-lasting antitumor immunity by the key process of immunogenic cell death (ICD). Furthermore, Off-target effects of chemotherapy on immune cell subsets mainly involve activation of immune effector cells including natural killer (NK) cells, dendritic cells (DCs), and cytotoxic T cells, and depletion of immunosuppressive cells including Treg cells, M2 macrophages and myeloid-derived suppressor cells (MDSCs). Current mini-review summarized recent large clinical trials regarding the combination of chemotherapy and immunotherapy in BC and addressed the molecular mechanisms of immunostimulatory properties of chemotherapy in BC. The purpose of our work was to explore the immune-stimulating effects of chemotherapy at the molecular level based on the evidence from clinical trials, which might be a rationale for combinations of chemotherapy and immunotherapy in BC.


2021 ◽  
pp. 1847-1852
Author(s):  
Kenju Ando ◽  
Akihiko Shimomura ◽  
Ryo Nasu ◽  
Misao Nakanishi ◽  
Yukino Kawamura ◽  
...  

Therapy-related myelodysplastic syndrome (tMDS) and acute myeloid leukemia (tAML) are lethal complications of chemotherapy. The incidence rates are expected to increase owing to improvements of cancer treatment. Early diagnosis of tMDS/AML is crucial because AML progresses rapidly. Hematopoietic stem cell transplantation (HSCT) is the only current treatment to prolong survival; however, patients with tMDS/AML are more likely to be intolerable to HSCT if they have other active solid tumors. An effective treatment for patients with tMDS/AML who are not candidates for HSCT is not established. We present a case of tAML that developed during chemotherapy for treating active ovarian cancer. The patient presented with thrombocytopenia that was initially suggested to be chemotherapy-induced thrombocytopenia. The patient was not a candidate for HSCT because of active cancer. However, she was able to receive azacitidine because her ovarian cancer responded well to chemotherapy. Pancytopenia is a common symptom of both chemotherapy-induced bone marrow suppression and tMDS/AML; thus, it may be difficult to distinguish between them at the first presentation. Given the prediction that the tMDS/AML incidence will increase as the survival of cancer patients improves, oncologists should be aware of the risks of tMDS/AML in patients with a history of cytotoxic chemotherapy. Although the indications for intensive care of tAML for patients with active solid tumors are poor, some patients might be able to receive cytotoxic treatment for tAML if the active solid tumors remain stable. Further studies focused on tMDS/AML with active solid tumors are needed to develop an effective treatment.


Antioxidants ◽  
2021 ◽  
Vol 11 (1) ◽  
pp. 35
Author(s):  
Joaquín Guerra ◽  
Jesús Devesa

Along with genetic mutations, aberrant epigenetic alterations are the initiators of head and neck cancer carcinogenesis. Currently, several drugs are being developed to correct these epigenetic alterations, known as epidrugs. Some compounds with an antioxidant effect have been shown to be effective in preventing these malignant lesions and in minimizing the complications derived from cytotoxic treatment. Furthermore, in vitro and in vivo studies show a promising role in the treatment of head and neck squamous cell carcinoma (HNSCC). This is the case of supplements with DNA methylation inhibitory function (DNMTi), such as epigallocatechin gallate, sulforaphane, and folic acid; histone deacetylase inhibitors (HDACi), such as sodium butyrate and melatonin or histone acetyltransferase inhibitors (HATi), such as curcumin. The objective of this review is to describe the role of some antioxidants and their epigenetic mechanism of action, with special emphasis on melatonin and butyric acid given their organic production, in the prevention and treatment of HNSCC.


2021 ◽  
Vol 11 ◽  
Author(s):  
Konstantinos Liapis ◽  
Ioannis Kotsianidis

Chronic myelomonocytic leukemia (CMML) is a rare clonal haematological malignancy bearing characteristics of both myelodysplastic syndromes and myeloproliferative neoplasms. It primarily affects older people (median age at diagnosis ~72 years). There are many challenges encountered in its treatment. One striking issue is the lack of strong clinical evidence from large randomized clinical trials for treating this disease. Another issue is that patients with CMML have highly variable outcomes with current treatments. Additional challenges include a wider application of current knowledge, an improved understanding of pathogenesis, development of new therapies, and management of refractory cases/disease progression. It is clear that there is still progress to be made. Here, we review the available first-line treatment options for advanced CMML. Emphasis has been placed on choosing between hypomethylating agents and cytotoxic treatments, on the basis on disease-specific and patient-specific characteristics. A proper selection between these two treatments could lead to a better quality of care for patients with CMML.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Ebun Omoyinmi ◽  
Dorota Rowczenio ◽  
Neil Sebire ◽  
Paul A. Brogan ◽  
Despina Eleftheriou

Abstract Background Mevalonate kinase deficiency (MKD) is a rare autoinflammatory condition caused by biallelic loss-of-function (LOF) mutations in mevalonate kinase (MVK) gene encoding the enzyme mevalonate kinase. Patients with MKD display a variety of non-specific clinical manifestations, which can lead to diagnostic delay. We report the case of a child presenting with vasculitis that was found by genetic testing to be caused by MKD, and now add this autoinflammatory disease to the ever-expanding list of causes of monogenic vasculitides. Case presentation A 2-year-old male presented with an acute 7-day history of high-grade fever, abdominal pain, diarrhoea, rectal bleeding and extensive purpuric and necrotic lesions, predominantly affecting the lower limbs. He had been suffering from recurrent episodes of fever from early in infancy, associated with maculopapular/petechial rashes triggered by intercurrent infection, and after vaccines. Extensive infection screen was negative. Skin biopsy revealed small vessel vasculitis. Visceral digital subtraction arteriography was normal. With a diagnosis of severe idiopathic cutaneous vasculitis, he was treated with corticosteroids and mycophenolate mofetil. Despite that his acute phase reactants remained elevated, fever persisted and the vasculitic lesions progressed. Next-generation sequencing revealed compound heterozygous mutation in MVK c.928G > A (p.V310M) and c.1129G > A (p.V377I) while reduced mevalonate enzyme activity was confirmed suggesting a diagnosis of MKD as a cause of the severe vasculitis. Prompt targeted treatment with IL-1 blockade was initiated preventing escalation to more toxic vasculitis therapies and reducing unnecessary exposure to cytotoxic treatment. Conclusions Our report highlights the broad clinical phenotype of MKD that includes severe cutaneous vasculitis and emphasizes the need to consider early genetic screening for young children presenting with vasculitis to exclude a monogenic vasculitis which may be amenable to targeted treatment.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2425-2425
Author(s):  
Paul M. Barr ◽  
Hongli Li ◽  
Brian K. Link ◽  
Christopher R. Flowers ◽  
Richard Burack ◽  
...  

Abstract While the majority of follicular lymphoma (FL) patients have an overall survival of nearly 2 decades, a subset of patients has a markedly inferior survival. Across randomized studies, 20% of patients will respond poorly to first-line chemoimmunotherapy and account largely for the early deaths in the larger FL population. This group represents the largest unmet need in FL, for which a precision approach to therapy must be developed. With the development of newer monoclonal antibodies, immunomodulatory agents, therapies targeting molecules downstream of the B-cell receptor and novel cellular strategies, non-cytotoxic treatment has the potential to improve outcomes for patients with early progressing FL. There are several validated clinical factors known to correlate with disease outcome in newly diagnosed FL including age, lactate dehydrogenase, β2-microglobulin and disease extent that have been incorporated in prognostic systems such as FLIPI and FLIPI2. More recently, genetic biomarkers have been identified, including MLL2, EZH2, IRF4, CREPPB, and EPHA7 which reflect the disease biology as well as the impact of the lymphoma microenvironment. The addition of these molecular aberrations to clinical factors has led to the development of the M7-FLIPI as well as a 23-gene score, improving risk prognostication for newly diagnosed FL patients. However, such systems have shown a limited ability to predict progression or relapse within 2 years of chemotherapy. As such, identification of these patients at diagnosis or prior to therapy is currently not possible. S1608 was developed to 1) enable identification of high-risk patients using clinical and molecular markers by validating the m7-FLIPI prognostic system and to 2) identify the novel therapeutic approaches most active in this population. This study is enrolling high-risk patients, refractory to chemoimmunotherapy, and in randomized fashion, comparing novel regimens against additional chemotherapy to identify the most active non-chemotherapeutic strategies for this population. Eligible patients must be 18 years or older with grade 1, 2 or 3a FL and have relapsed or progressed with 2 years of finishing their first course of chemoimmunotherapy. Previous chemotherapy must have been CHOP or bendamustine based. Patients are eligible regardless of anti-CD20 therapy used, whether radiation therapy had been administered and whether or not maintenance therapy was utilized. Note that patients are required to have evidence of progressive disease within 2 years but do not have to be registered within 2 years. These high-risk patients are randomized to 12 months of lenalidomide, umbralisib or additional chemotherapy (for 6 months), all combined with 12 months of obinutuzumab. The primary clinical endpoint is CR rate after 6 cycles, allowing responding patient to proceed with consolidative cellular therapies if desired by the treating physician. Biopsies from diagnosis and at the time of relapse as well as circulating tumor DNA are being collected to prospectively evaluate the m7-FLIPI and to identify additional predictive markers. S1608 is a collaborative effort amongst the SWOG, Alliance and ECOG-ACRIN cooperative groups. The study represents one of the only prospective efforts to characterize early progressing FL and the only randomized trial comparing treatment strategies for this group of follicular lymphoma patients most in need of alternative therapies. Funding: NIH/NCI/NCTN grants U10CA180888, U10CA180819, U10CA180820, U10CA180821; and TG Therapeutics, Inc. Figure 1 Figure 1. Disclosures Barr: Genentech: Consultancy; AstraZeneca: Consultancy; Morphosys: Consultancy; TG Therapeutics: Consultancy; Beigene: Consultancy; Abbvie/Pharmacyclics: Consultancy; Bristol Meyers Squibb: Consultancy; Seattle Genetics: Consultancy; Janssen: Consultancy; Gilead: Consultancy. Link: Novartis, Jannsen: Research Funding; MEI: Consultancy; Genentech/Roche: Consultancy, Research Funding. Flowers: Cellectis: Research Funding; Nektar: Research Funding; Takeda: Research Funding; TG Therapeutics: Research Funding; BeiGene: Consultancy; 4D: Research Funding; Karyopharm: Consultancy; Morphosys: Research Funding; Guardant: Research Funding; Bayer: Consultancy, Research Funding; Genmab: Consultancy; Eastern Cooperative Oncology Group: Research Funding; SeaGen: Consultancy; Genentech/Roche: Consultancy, Research Funding; Pharmacyclics/Janssen: Consultancy; Burroughs Wellcome Fund: Research Funding; AbbVie: Consultancy, Research Funding; Adaptimmune: Research Funding; Janssen: Research Funding; Iovance: Research Funding; Acerta: Research Funding; Kite: Research Funding; Allogene: Research Funding; EMD: Research Funding; Amgen: Research Funding; Celgene: Consultancy, Research Funding; Ziopharm: Research Funding; Novartis: Research Funding; Pfizer: Research Funding; Sanofi: Research Funding; National Cancer Institute: Research Funding; Xencor: Research Funding; Spectrum: Consultancy; Gilead: Consultancy, Research Funding; Epizyme, Inc.: Consultancy; Biopharma: Consultancy; Denovo: Consultancy; Cancer Prevention and Research Institute of Texas: CPRIT Scholar in Cancer Research: Research Funding; Pharmacyclics: Research Funding. Weigert: Janssen: Speakers Bureau; Epizyme: Membership on an entity's Board of Directors or advisory committees; Roche: Research Funding. Herrera: ADC Therapeutics: Consultancy, Research Funding; Tubulis: Consultancy; Karyopharm: Consultancy; Kite, a Gilead Company: Research Funding; Bristol Myers Squibb: Consultancy, Research Funding; Gilead Sciences: Research Funding; Merck: Consultancy, Research Funding; Genentech: Consultancy, Research Funding; Takeda: Consultancy; Seagen: Consultancy, Research Funding; AstraZeneca: Consultancy, Research Funding. Weinstock: SecuraBio: Consultancy; ASELL: Consultancy; Bantam: Consultancy; Abcuro: Research Funding; Verastem: Research Funding; Daiichi Sankyo: Consultancy, Research Funding; AstraZeneca: Consultancy; Travera: Other: Founder/Equity; Ajax: Other: Founder/Equity. Leonard: ADC Therapeutics, AstraZeneca, Bayer, BMS/Celgene, Epizyme, Inc., Genmab, Gilead/Kite, Karyopharm, BMS/Celgene, Regeneron, MEI Pharma, Miltenyi, Roche/Genentech, Sutro: Consultancy; Roche/Genentech: Consultancy. Kahl: Abbvie, BeiGene, AstraZeneca, Acerta: Research Funding; Research to Practice: Speakers Bureau; Abbvie, ADCT, AstraZeneca, Beigene, Celgene, Teva, Janssen, MTEM, Bayer, InCyte, Adaptive, Genentech, Roche, MEI, KITE, TG Therapeutics, Epizyme, Takeda: Consultancy. Smith: Celgene, Genetech, AbbVie: Consultancy; Alexion, AstraZeneca Rare Disease: Other: Study investigator. Friedberg: Novartis: Other: DSMC ; Acerta: Other: DSMC ; Bayer: Other: DSMC .


2021 ◽  
Author(s):  
Kai-Lin Peng ◽  
Harish N. Vasudevan ◽  
Dennis T. Lockney ◽  
Rachel Baum ◽  
Ronald C. Hendrickson ◽  
...  

AbstractMedulloblastoma is a pediatric brain tumor arising from the cerebellum and brainstem that is curable with surgery and aggressive cytotoxic therapy including craniospinal irradiation and chemotherapy. Preclinical models indicate that a small pool of de-differentiated, stem cell-like medulloblastoma cells are resistant to cytotoxic treatment and contribute to medulloblastoma relapse after aggressive therapy. Here, we identify Miat as a Shh and Myc regulated long noncoding RNA (lncRNA) that is required for maintenance of a treatment-resistant medulloblastoma stem-like phenotype. Loss of Miat delays medulloblastoma formation in genetically defined mouse models of Shh medulloblastoma and enforces differentiation of tumorigenic stem-like medulloblastoma cells into a non-tumorigenic cell state. Miat facilitates treatment resistance in part by downregulating p53 signaling and impairing radiation induced cell death in stem-like MB cells, which can be reversed by therapeutic inhibition of Miat with antisense oligonucleotides. The RNA binding protein Metadhedrin (Mtdh), which is associated with resistance to cytotoxic therapy in numerous types of cancer, co-localizes with Miat in stem-like medulloblastoma cells. Further, loss of Mtdh activates p53 signaling and reduces tumorigenicity in stem-like medulloblastoma cells. Taken together, these data reveal a critical role for the lncRNA Miat in sustaining a treatment resistant pool of tumorigenic stem-like medulloblastoma cells.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Oriol Pich ◽  
Albert Cortes-Bullich ◽  
Ferran Muiños ◽  
Marta Pratcorona ◽  
Abel Gonzalez-Perez ◽  
...  

AbstractChemotherapies may increase mutagenesis of healthy cells and change the selective pressures in tissues, thus influencing their evolution. However, their contributions to the mutation burden and clonal expansions of healthy somatic tissues are not clear. Here, exploiting the mutational footprint of some chemotherapies, we explore their influence on the evolution of hematopoietic cells. Cells of Acute Myeloid Leukemia (AML) secondary to treatment with platinum-based drugs show the mutational footprint of these drugs, indicating that non-malignant blood cells receive chemotherapy mutations. No trace of the 5-fluorouracil (5FU) mutational signature is found in AMLs secondary to exposure to 5FU, suggesting that cells establishing the leukemia could be quiescent during treatment. Using the platinum-based mutational signature as a barcode, we determine that the clonal expansion originating the secondary AMLs begins after the start of the cytotoxic treatment. Its absence in clonal hematopoiesis cases is consistent with the start of the clonal expansion predating the exposure to platinum-based drugs.


2021 ◽  
Author(s):  
Ulrik N Lassen ◽  
Lydia E Makaroff ◽  
Albrecht Stenzinger ◽  
Antoine Italiano ◽  
Gilles Vassal ◽  
...  

Molecular characterization of tumors has shifted cancer treatment strategies away from nonspecific cytotoxic treatment of histology-specific tumors toward targeting of actionable mutations that can be found across multiple cancer types. The development of high-throughput technologies such as next-generation sequencing, combined with decision support applications and availability of patient databases, has provided tools that optimize disease management. Precision oncology has proven success in improving outcomes and quality of life, as well as identifying and overcoming mechanisms of drug resistance and relapse. Addressing challenges that impede its use will improve matching of therapies to patients. Here we review the current status of precision oncology medicine, emphasizing its impact on patients – what they understand about precision oncology medicine and their hopes for the future.


Andrologia ◽  
2021 ◽  
Author(s):  
Yi‐ting Cai ◽  
Cheng‐liang Xiong ◽  
Tian‐shu Liu ◽  
Shi‐liang Shen ◽  
Jin‐peng Rao ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document