Journal articles on the topic 'Targeted therapy of hematological malignancie'

To see the other types of publications on this topic, follow the link: Targeted therapy of hematological malignancie.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Targeted therapy of hematological malignancie.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Armitage, James O. "Targeted therapy and hematological malignancy." Targeted Oncology 4, no. 1 (February 14, 2009): 1–2. http://dx.doi.org/10.1007/s11523-008-0098-1.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Kachlany, Scott C., Amy Le, and Benjamin A. Belinka. "Leukotoxin (Leukothera™), a Targeted Therapy for Hematological Malignancies." Blood 116, no. 21 (November 19, 2010): 3284. http://dx.doi.org/10.1182/blood.v116.21.3284.3284.

Full text
Abstract:
Abstract Abstract 3284 Leukotoxin (Leukothera™) is a bacterial protein toxin that naturally targets and kills disease associated white blood cells (WBCs) expressing the activated form of leukocyte function antigen-1 (LFA-1). While leukotoxin has minimal effects on resting and healthy cells, it causes significant death of malignant WBCs associated with leukemias and lymphomas. Leukotoxin is a unique biologic in that it already provides both toxicity and specificity without requiring fusion of the protein to other molecules such as antibody fragments or cytokines. In vivo efficacy was demonstrated in an HL-60 SCID mouse leukemia model previously. In the present work, we compared leukotoxin to the standard chemotherapeutic agents, doxorubicin and cytarabine in a SCID mouse model using THP-1 leukemia cells. Mice (n=14 per group) were injected with THP-1 cells intravenously (i.v.) and then administered either leukotoxin or standard agents i.v. Mice received three doses, once daily, of leukotoxin (1.5 mg/kg) or five doses of doxorubicin (0.5 mg/kg) or cytarabine (10 mg/kg) once daily. Leukemic mice that were treated with leukotoxin maintained and increased body weight more effectively than those which received vehicle alone or the standard agents. The leukotoxin-treated mice showed significantly (p<0.001) higher mean survival than mice treated with vehicle or cytarabine over the 60-day observation period. Necropsy and histopathology of animals after the 60-day period revealed that test animals treated with vehicle or standard agents developed internal tumors on organs such as the thymus and lymph nodes. In contrast, none of the leukotoxin-treated mice developed internal tumors. To determine if leukotoxin could block migration of malignant cells, and thus formation of internal tumors, we performed a cellular migration assay using activated monocytes and human brain endothelial cells. It was found that even a low dose of leukotoxin (10 ng/ml) caused significant suppression (>80%) of monocyte migration across an endothelial barrier. Hence, leukotoxin has the potential to not only deplete malignant WBCs, but may also prevent their spread to other tissues. To test the general safety of leukotoxin, mice were injected with 1 mg/kg for three weeks. None of the mice showed signs of illness or changes in normal behavior and all continued to gain weight throughout the study period. In addition, to determine if a neutralizing antibody response to leukotoxin was generated, we challenged mice with high doses of leukotoxin and assayed monoclonal antibody from ten independent hybridomas. Results showed that antibody was able to bind to and recognize leukotoxin, but did not cause significant neutralization in a bioassay. In conclusion, leukotoxin may represent a highly effective and safe option for patients with hematologic malignancies, especially those with relapsed and refractory disease. Disclosures: Kachlany: Actinobac Biomed, Inc.: Consultancy, Equity Ownership. Belinka:Actinobac Biomed, Inc.: Employment, Equity Ownership.
APA, Harvard, Vancouver, ISO, and other styles
3

Joshi, Dolly, Kanjaksha Gosh, and Babu Rao Vundinti. "MicroRNAs in hematological malignancies: a novel approach to targeted therapy." Hematology 17, no. 3 (May 2012): 170–75. http://dx.doi.org/10.1179/102453312x13376952196656.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Ma, Haiqing, Saradhi Mallampati, Gang An, and Jin Wang. "Targeted Therapy in Hematological Malignancies: From Basic Research to Clinical Practice." BioMed Research International 2015 (2015): 1–2. http://dx.doi.org/10.1155/2015/157570.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Podhorecka, Monika, Justyna Markowicz, Agnieszka Szymczyk, and Johannes Pawlowski. "Target Therapy in Hematological Malignances: New Monoclonal Antibodies." International Scholarly Research Notices 2014 (October 30, 2014): 1–16. http://dx.doi.org/10.1155/2014/701493.

Full text
Abstract:
Apart from radio- and chemotherapy, monoclonal antibodies (MoAbs) represent a new, more selective tool in the treatment of hematological malignancies. MoAbs bind with the specific antigens of the tumors. This interaction is a basis for targeted therapies which exhibit few side effects and significant antitumor activity. This review provides an overview of the functional characteristics of MoAbs, with some examples of their clinical application. The promising results in the treatment of hematological malignancies have led to the more frequent usage of MoAbs in the therapy. Development of MoAbs is a subject of extensive research. They are a promising method of cancer treatment in the future.
APA, Harvard, Vancouver, ISO, and other styles
6

Liang, Xuewu, Hong Liu, and Yingjie Zhang. "Novel-targeted therapy for hematological malignancies with JAK and HDAC dual inhibitors." Future Medicinal Chemistry 11, no. 15 (August 2019): 1849–52. http://dx.doi.org/10.4155/fmc-2019-0168.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Leni, Zaira, Geetha Parakkal, and Alexandre Arcaro. "Emerging Metabolic Targets in the Therapy of Hematological Malignancies." BioMed Research International 2013 (2013): 1–12. http://dx.doi.org/10.1155/2013/946206.

Full text
Abstract:
During the last decade, the development of anticancer therapies has focused on targeting neoplastic-related metabolism. Cancer cells display a variety of changes in their metabolism, which enable them to satisfy the high bioenergetic and biosynthetic demands for rapid cell division. One of the crucial alterations is referred to as the “Warburg effect”, which involves a metabolic shift from oxidative phosphorylation towards the less efficient glycolysis, independent of the presence of oxygen. Although there are many examples of solid tumors having altered metabolism with high rates of glucose uptake and glycolysis, it was only recently reported that this phenomenon occurs in hematological malignancies. This review presents evidence that targeting the glycolytic pathway at different levels in hematological malignancies can inhibit cancer cell proliferation by restoring normal metabolic conditions. However, to achieve cancer regression, high concentrations of glycolytic inhibitors are used due to limited solubility and biodistribution, which may result in toxicity. Besides using these inhibitors as monotherapies, combinatorial approaches using standard chemotherapeutic agents could display enhanced efficacy at eradicating malignant cells. The identification of the metabolic enzymes critical for hematological cancer cell proliferation and survival appears to be an interesting new approach for the targeted therapy of hematological malignancies.
APA, Harvard, Vancouver, ISO, and other styles
8

Gao, Rili, Yikai Zhang, Chengwu Zeng, and Yangqiu Li. "The role of NFAT in the pathogenesis and targeted therapy of hematological malignancies." European Journal of Pharmacology 921 (April 2022): 174889. http://dx.doi.org/10.1016/j.ejphar.2022.174889.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Kim, Miyoung, Jane Snowdon, S. Dilhan Weeraratne, Winnie Felix, Lionel Lim, Irene Dankwa-Mullan, Young Kyung Lee, et al. "Clinical insights for hematological malignancies from an artificial intelligence decision-support tool." Journal of Clinical Oncology 37, no. 15_suppl (May 20, 2019): e13023-e13023. http://dx.doi.org/10.1200/jco.2019.37.15_suppl.e13023.

Full text
Abstract:
e13023 Background: Next generation sequencing (NGS) in hematological tumors is increasingly shaping clinical treatment decisions at the point of care. While the impact of NGS panels in solid tumors is largely therapeutic, targeted sequencing in hematological tumors can additionally provide diagnostic and prognostic insights. Additional data generated in hematological tumor sequencing makes manual interpretation and annotation of variants tedious and non-scalable. In this study we compared hematological tumor variant interpretation using an artificial intelligence decision-support system, Watsonä for Genomics (WfG), with expert guided manual curation. Methods: Patients with hematological tumors at Hallym University, College of Medicine between December 2017 and December 2018, were sequenced using the 54 gene Illumina TruSight Myeloid Panel. WfG interpreted and annotated all patients’ sequencing results, a subset of which were assessed manually to ascertain concordance. Results: 54 South Korean patients with hematological malignancies were analyzed (23 Acute Myeloid Leukemia, 12 myeloproliferative neoplasm, 5 myelodysplastic syndrome, 5 multiple myeloma and 9 others). Comparison of manual and WfG interpretation of 10 randomly selected cases yielded 90% (9/10) concordance and identification of 9 clinically actionable variants (33%) not found in manual interpretation. In total, WfG identified that 71% (38/54) of all cases had at least one clinically actionable therapeutic alteration (a variant targeted by a US FDA approved drug, off-label drug, or clinical trial). 33% (18/54) of cases had genes that were targeted by a US FDA approved therapy including JAK2, IDH1, IDH2, and FLT3. In cases without therapeutic alterations, WfG identified diagnostic or prognostic insights in an additional 20% (11/54) of patients. 9% (5/54) had no clinically actionable information. Conclusions: WfG variant interpretation correlated well with manually curated expert opinion and identified clinically actionable insights missed by manual interpretation. WfG has obviated the need for labor-intensive manual curation of clinical trials and therapy, enabling our center to exponentially scale our NGS operations.
APA, Harvard, Vancouver, ISO, and other styles
10

Miloudi, Hadjer, Vincent Camus, Antoine Taly, Brigitte Sola, and Fabrice Jardin. "Exportin 1 (or XPO1) abnormalities in hematological malignancies: from the gene to targeted therapy." Hématologie 23, no. 1 (January 2017): 43–56. http://dx.doi.org/10.1684/hma.2017.1208.

Full text
APA, Harvard, Vancouver, ISO, and other styles
11

Shakil, Md Salman, Mahruba Sultana Niloy, Kazi Mustafa Mahmud, Mohammad Amjad Kamal, and Md Asiful Islam. "Theranostic Potentials of Gold Nanomaterials in Hematological Malignancies." Cancers 14, no. 13 (June 21, 2022): 3047. http://dx.doi.org/10.3390/cancers14133047.

Full text
Abstract:
Hematological malignancies (HMs) are a heterogeneous group of blood neoplasia generally characterized by abnormal blood-cell production. Detection of HMs-specific molecular biomarkers (e.g., surface antigens, nucleic acid, and proteomic biomarkers) is crucial in determining clinical states and monitoring disease progression. Early diagnosis of HMs, followed by an effective treatment, can remarkably extend overall survival of patients. However, traditional and advanced HMs’ diagnostic strategies still lack selectivity and sensitivity. More importantly, commercially available chemotherapeutic drugs are losing their efficacy due to adverse effects, and many patients develop resistance against these drugs. To overcome these limitations, the development of novel potent and reliable theranostic agents is urgently needed to diagnose and combat HMs at an early stage. Recently, gold nanomaterials (GNMs) have shown promise in the diagnosis and treatment of HMs. Magnetic resonance and the surface-plasmon-resonance properties of GNMs have made them a suitable candidate in the diagnosis of HMs via magnetic-resonance imaging and colorimetric or electrochemical sensing of cancer-specific biomarkers. Furthermore, GNMs-based photodynamic therapy, photothermal therapy, radiation therapy, and targeted drug delivery enhanced the selectivity and efficacy of anticancer drugs or drug candidates. Therefore, surface-tuned GNMs could be used as sensitive, reliable, and accurate early HMs, metastatic HMs, and MRD-detection tools, as well as selective, potent anticancer agents. However, GNMs may induce endothelial leakage to exacerbate cancer metastasis. Studies using clinical patient samples, patient-derived HMs models, or healthy-animal models could give a precise idea about their theranostic potential as well as biocompatibility. The present review will investigate the theranostic potential of vectorized GNMs in HMs and future challenges before clinical theranostic applications in HMs.
APA, Harvard, Vancouver, ISO, and other styles
12

Xie, Bailu, Zhengdong Li, Jianfeng Zhou, and Wen Wang. "Current Status and Perspectives of Dual-Targeting Chimeric Antigen Receptor T-Cell Therapy for the Treatment of Hematological Malignancies." Cancers 14, no. 13 (June 30, 2022): 3230. http://dx.doi.org/10.3390/cancers14133230.

Full text
Abstract:
Single-targeted chimeric antigen receptor (CAR) T cells tremendously improve outcomes for patients with relapsed/refractory hematological malignancies and are considered a breakthrough therapy. However, over half of treated patients experience relapse or refractory disease, with antigen escape being one of the main contributing mechanisms. Dual-targeting CAR T-cell therapy is being developed to minimize the risk of relapse or refractory disease. Preclinical and clinical data on five categories of dual-targeting CAR T-cell therapies and approximately fifty studies were summarized to offer insights and support the development of dual-targeting CAR T-cell therapy for hematological malignancies. The clinical efficacy (durability and survival) is validated and the safety profiles of dual-targeting CAR T-cell therapy are acceptable, although there is still room for improvement in the bispecific CAR structure. It is one of the best approaches to optimize the bispecific CAR structure by boosting T-cell transduction efficiency and leveraging evidence from preclinical activity and clinical efficacy.
APA, Harvard, Vancouver, ISO, and other styles
13

Ahmed, Shah Newaz, and Arun Kumar. "CAR T-cell therapy-transforming treatment of hematological malignancies." Asian Journal of Medical Sciences 14, no. 2 (February 1, 2023): 1–2. http://dx.doi.org/10.3126/ajms.v14i2.50899.

Full text
Abstract:
Hematological malignancies are a significant cause of mortality and morbidity in all age groups. According to the Global Burden of Disease Cancer Collaboration, there were about one million incident cases of hematological malignancies worldwide in 2016. In an analysis of NORDCAN database, it was observed that the 5-year survival rates during the period 1999–2003 for Hodgkin lymphoma, Non-Hodgkin lymphoma, acute leukemia, other leukemia, and multiple myeloma were 80, 50–60, 38–49, 60–73, and 28–41%, respectively. The treatment of these blood cancers has seen rapid advances in modern medicine. However, mortality is still very high and globally, about half a million deaths were attributed to these malignancies in 2016 alone.1 In some cancers, such as the relapsed/refractory B-cell precursor-ALL, the 5-year survival rate is only 10% with conventional treatment. Conventionally, chemotherapy has been the mainstay of treatment supported by radiation therapy, targeted therapy, or stem-cell transplantation resulting in improved outcomes. Novel treatment options that improve outcomes are under research worldwide. The chimeric antigen receptor T-cell therapy (CAR-T) is a promising novel anticancer treatment modality. A CAR-T therapy consists of infusion of engineered T-cells that express a chimeric antigen receptor on their cell membrane. In-vivo, CAR-T cells eliminate tumor cells with precision by interacting with the tumor-associated antigens (TAAs) on tumor cell surface independent of the expression of major histocompatibility complex. CAR is a recombinant immunoreceptor composed of an extracellular binding domain (TAA-reactive antibody derived Single-chain variable fragments), a hinge region, a transmembrane domain, and one or more intracellular signalling domains (CD3ζchain). Second-, third-, and fourth-generations of these receptors have costimulatory domains such as CD28 and/or 4-1BB which enhance auto-proliferation, cytokine secretion, apoptosis resistance, and in-vivo persistence. Fourth generation CARs have additional enzyme secreting properties which degrade extracellular matrix of solid tumours. The first FDA approved therapies, tisagenlecleucel followed by axicabtagene ciloleucel, are third generation anti-CD19 CAR T-cells indicated for treatment of refractory B-cell acute lymphoblastic leukemia (in children and young adults) and refractory diffuse large B-cell lymphoma (in adults), respectively. The approval was based on Phase II trials (ELIANA trial for tisagenlecleucel and ZUMA-1 trial for axicabtagene). An objective response rate of 81 and 82% was observed, respectively. The success of these therapies has rekindled the hope for targeting other malignancies which have been hitherto refractory to treatment. At present, CAR-T cell therapy targeting CD7/CD23/ CD33/CD34/CD38/CD56/CD117/CD123/CD133/ Mucl (for acute myeloid leukemia), CD19/CD20/CD22/ EGFR (for chronic lymphocytic leukemia), CD30 (for Hodgkin lymphoma), and CD38/CD138 (for multiple myeloma) are in various stages of Phase I/II clinical trials (nearly 150). Despite the initial better outcomes, it is too early to predict the final picture. Serious adverse drug reaction (CRS-cytokine release syndrome and NT-neurotoxicity) leading to fatality has already been reported. Moreover, concerns have been expressed about tumor relapse, genotoxicity, and autoimmunity as long-term consequences. In the Phase 2 ELARA trial, tisagenlecleucel was found to be safe and effective in adult relapsed or refractory follicular lymphoma. However, the BELINDA trial (Phase III trial) failed to show superiority to standard salvage therapy. On an optimistic note, ZUMA-7 trial and TRANSFORM trial for axicabtagene in early relapsed or refractory large B-cell lymphoma demonstrated improved survival and response outcomes compared to standard therapy. A handful of Phase 3 trials are underway to test the efficacy and safety of the “revolutionary” drugs in the treatment of hematological malignancies. The acceptability of these “living drugs” in the future will depend on the results of these trials and beyond.
APA, Harvard, Vancouver, ISO, and other styles
14

Volkov, Dmitry V., George V. Tetz, Yury P. Rubtsov, Alexey V. Stepanov, and Alexander G. Gabibov. "Neutrophil Extracellular Traps (NETs): Opportunities for Targeted Therapy." Acta Naturae 13, no. 3 (November 15, 2021): 15–23. http://dx.doi.org/10.32607/actanaturae.11503.

Full text
Abstract:
Antitumor therapy, including adoptive immunotherapy, inevitably faces powerful counteraction from advanced cancer. If hematological malignancies are currently amenable to therapy with CAR-T lymphocytes (T-cells modified by the chimeric antigen receptor), solid tumors, unfortunately, show a significantly higher degree of resistance to this type of therapy. As recent studies show, the leading role in the escape of solid tumors from the cytotoxic activity of immune cells belongs to the tumor microenvironment (TME). TME consists of several types of cells, including neutrophils, the most numerous cells of the immune system. Recent studies show that the development of the tumor and its ability to metastasize directly affect the extracellular traps of neutrophils (neutrophil extracellular traps, NETs) formed as a result of the response to tumor stimuli. In addition, the nuclear DNA of neutrophils the main component of NETs erects a spatial barrier to the interaction of CAR-T with tumor cells. Previous studies have demonstrated the promising potential of deoxyribonuclease I (DNase I) in the destruction of NETs. In this regard, the use of eukaryotic deoxyribonuclease I (DNase I) is promising in the effort to increase the efficiency of CAR-T by reducing the NETs influence in TME. We will examine the role of NETs in TME and the various approaches in the effort to reduce the effect of NETs on a tumor.
APA, Harvard, Vancouver, ISO, and other styles
15

Karvonen, Hanna, Wilhelmiina Niininen, Astrid Murumägi, and Daniela Ungureanu. "Targeting ROR1 identifies new treatment strategies in hematological cancers." Biochemical Society Transactions 45, no. 2 (April 13, 2017): 457–64. http://dx.doi.org/10.1042/bst20160272.

Full text
Abstract:
Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a member of the ROR receptor family consisting of two closely related type I transmembrane proteins ROR1 and ROR2. Owing to mutations in their canonical motifs required for proper kinase activity, RORs are classified as pseudokinases lacking detectable catalytic activity. ROR1 stands out for its selective and high expression in numerous blood and solid malignancies compared with a minimal expression in healthy adult tissues, suggesting high potential for this molecule as a drug target for cancer therapy. Current understanding attributes a survival role for ROR1 in cancer cells; however, its oncogenic function is cancer-type-specific and involves various signaling pathways. High interest in ROR1-targeted therapies resulted in the development of ROR1 monoclonal antibodies such as cirmtuzumab, currently in a phase I clinical trial for chronic lymphocytic leukemia. Despite these advances in translational studies, the molecular mechanism employed by ROR1 in different cancers is not yet fully understood; therefore, more insights into the oncogenic role of ROR1 signaling are crucial in order to optimize the use of targeted drugs. Recent studies provided evidence that targeting ROR1 simultaneously with inhibition of B-cell receptor (BCR) signaling is more effective in killing ROR1-positive leukemia cells, suggesting a synergistic correlation between co-targeting ROR1 and BCR pathways. Although this synergy has been previously reported for B-cell acute lymphoblastic leukemia, the molecular mechanism appears rather different. These results provide more insights into ROR1–BCR combinatorial treatment strategies in hematological malignancies, which could benefit in tailoring more effective targeted therapies in other ROR1-positive cancers.
APA, Harvard, Vancouver, ISO, and other styles
16

Garcia, Anita A. "Small Molecules: Big Changes in the Cancer Treatment Paradigm." Journal of Pharmacy Practice 21, no. 1 (February 2008): 17–35. http://dx.doi.org/10.1177/0897190008314779.

Full text
Abstract:
Small molecules, a growing class of targeted therapies, have flourished over the last decade. With increased knowledge on molecular cell signaling, targeted therapy has been refined to targeting molecular targets upstream from the nucleus that are key players in the communication system that regulates cancer cell growth. This article reviews the mechanisms of small molecules with a particular emphasis on tyrosine kinase inhibitors, as well as the literature that supports the current clinical use in the treatment of a variety of solid and hematological malignancies.
APA, Harvard, Vancouver, ISO, and other styles
17

Zhang, Jianxiang, and Lingyu Wang. "The Emerging World of TCR-T Cell Trials Against Cancer: A Systematic Review." Technology in Cancer Research & Treatment 18 (January 1, 2019): 153303381983106. http://dx.doi.org/10.1177/1533033819831068.

Full text
Abstract:
T-cell receptor–engineered T-cell therapy and chimeric antigen receptor T-cell therapy are 2 types of adoptive T-cell therapy that genetically modify natural T cells to treat cancers. Although chimeric antigen receptor T-cell therapy has yielded remarkable efficacy for hematological malignancies of the B-cell lineages, most solid tumors fail to respond significantly to chimeric antigen receptor T cells. T-cell receptor–engineered T-cell therapy, on the other hand, has shown unprecedented promise in treating solid tumors and has attracted growing interest. In order to create an unbiased, comprehensive, and scientific report for this fast-moving field, we carefully analyzed all 84 clinical trials using T-cell receptor–engineered T-cell therapy and downloaded from ClinicalTrials.gov updated by June 11, 2018. Informative features and trends were observed in these clinical trials. The number of trials initiated each year is increasing as expected, but an interesting pattern is observed. NY-ESO-1, as the most targeted antigen type, is the target of 31 clinical trials; melanoma is the most targeted cancer type and is the target of 33 clinical trials. Novel antigens and underrepresented cancers remain to be targeted in future studies and clinical trials. Unlike chimeric antigen receptor T-cell therapy, only about 16% of the 84 clinical trials target against hematological malignancies, consistent with T-cell receptor–engineered T-cell therapy’s high potential for solid tumors. Six pharma/biotech companies with novel T-cell receptor–engineered T-cell ideas and products were examined in this review. Multiple approaches have been utilized in these companies to increase the T-cell receptor’s affinity and efficiency and to minimize cross-reactivity. The major challenges in the development of the T-cell receptor–engineered T-cell therapy due to tumor microenvironment were also discussed here.
APA, Harvard, Vancouver, ISO, and other styles
18

Ye, Baixin, Creed M. Stary, Qingping Gao, Qiongyu Wang, Zhi Zeng, Zhihong Jian, Lijuan Gu, and Xiaoxing Xiong. "Genetically Modified T-Cell-Based Adoptive Immunotherapy in Hematological Malignancies." Journal of Immunology Research 2017 (2017): 1–13. http://dx.doi.org/10.1155/2017/5210459.

Full text
Abstract:
A significant proportion of hematological malignancies remain limited in treatment options. Immune system modulation serves as a promising therapeutic approach to eliminate malignant cells. Cytotoxic T lymphocytes (CTLs) play a central role in antitumor immunity; unfortunately, nonspecific approaches for targeted recognition of tumor cells by CTLs to mediate tumor immune evasion in hematological malignancies imply multiple mechanisms, which may or may not be clinically relevant. Recently, genetically modified T-cell-based adoptive immunotherapy approaches, including chimeric antigen receptor (CAR) T-cell therapy and engineered T-cell receptor (TCR) T-cell therapy, promise to overcome immune evasion by redirecting the specificity of CTLs to tumor cells. In clinic trials, CAR-T-cell- and TCR-T-cell-based adoptive immunotherapy have produced encouraging clinical outcomes, thereby demonstrating their therapeutic potential in mitigating tumor development. The purpose of the present review is to (1) provide a detailed overview of the multiple mechanisms for immune evasion related with T-cell-based therapies; (2) provide a current summary of the applications of CAR-T-cell- as well as neoantigen-specific TCR-T-cell-based adoptive immunotherapy and routes taken to overcome immune evasion; and (3) evaluate alternative approaches targeting immune evasionviaoptimization of CAR-T and TCR-T-cell immunotherapies.
APA, Harvard, Vancouver, ISO, and other styles
19

Xiang, Wei, Yi Hui Lam, Giridharan Periyasamy, and Charles Chuah. "Application of High Throughput Technologies in the Development of Acute Myeloid Leukemia Therapy: Challenges and Progress." International Journal of Molecular Sciences 23, no. 5 (March 5, 2022): 2863. http://dx.doi.org/10.3390/ijms23052863.

Full text
Abstract:
Acute myeloid leukemia (AML) is a complex hematological malignancy characterized by extensive heterogeneity in genetics, response to therapy and long-term outcomes, making it a prototype example of development for personalized medicine. Given the accessibility to hematologic malignancy patient samples and recent advances in high-throughput technologies, large amounts of biological data that are clinically relevant for diagnosis, risk stratification and targeted drug development have been generated. Recent studies highlight the potential of implementing genomic-based and phenotypic-based screens in clinics to improve survival in patients with refractory AML. In this review, we will discuss successful applications as well as challenges of most up-to-date high-throughput technologies, including artificial intelligence (AI) approaches, in the development of personalized medicine for AML, and recent clinical studies for evaluating the utility of integrating genomics-guided and drug sensitivity testing-guided treatment approaches for AML patients.
APA, Harvard, Vancouver, ISO, and other styles
20

Wach, Małgorzata, Monika Podhorecka, Maria Cioch, Iwona Hus, Ewa Wąsik-Szczepanek, Bożena Sokołowska, and Marek Hus. "Advances in hematology – research that revolutionized patient care." Polish Journal of Public Health 125, no. 1 (March 1, 2015): 32–35. http://dx.doi.org/10.1515/pjph-2015-0019.

Full text
Abstract:
AbstractIn the last decades, substantial strides have been made in the diagnosis, treatment, and prevention of blood diseases. The new drugs to be used in combination with cytostatic therapy have been developed, based on increased understanding of the biology of neoplasia. The diagnosis of several diseases is based exclusively on cytogenetic and molecular analysis which has become a part of routine diagnostic management. Moreover, molecular definition has allowed the introduction of therapy targeted at molecular change characteristic for a given disease. The introduction of novel agents for the treatment of hematological disorders has resulted in a great improvement in response rate and median survival.The aim of this study is to show advances and possible future directions in the treatment of chosen hematological malignancies during the recent decades.
APA, Harvard, Vancouver, ISO, and other styles
21

Daraskevicius, Justinas, Vidmantas Petraitis, Linas Davainis, and Andrius Zucenka. "The Feasibility of Ibrexafungerp for the Treatment of Fungal Infections in Patients with Hematological Malignancies." Journal of Fungi 8, no. 5 (April 23, 2022): 440. http://dx.doi.org/10.3390/jof8050440.

Full text
Abstract:
Invasive fungal diseases (IFD) remain a major cause of morbidity and mortality in hematological patients, especially those undergoing hematopoietic stem cell transplantation (HSCT). Despite relatively high incidence, diagnosis and treatment remain a challenge due to non-specific manifestation and limited antifungal armamentarium. A first-in-class triterpenoid antifungal ibrexafungerp that acts by inhibiting the glucan synthase enzyme in the fungal cell wall was recently approved by the USA Food and Drug Administration for the treatment of vaginal yeast infections. Preclinical data show activity against numerous fungi species, including azole- and echinocandin-resistant strains. Preliminary data from ongoing phase 3 studies in IFD have been encouraging, but the role of ibrexafungerp in hematological patients who develop fungal infections has not yet been described. Herein, we discuss the feasibility of oral ibrexafungerp-based antifungal therapy for adult patients with hematological malignancies who have either undergone HSCT or received treatment with a novel targeted therapy agent. We present four clinical cases where ibrexafungerp alone or in combination with other antifungal agents was successfully employed for the management of refractory fungal infection. We describe real-life experiences showing the potential clinical implementation of ibrexafungerp for patients with hematological malignancies for the first time, and provoke future discussion.
APA, Harvard, Vancouver, ISO, and other styles
22

Roex, Gils, Tom Feys, Yves Beguin, Tessa Kerre, Xavier Poiré, Philippe Lewalle, Peter Vandenberghe, Dominique Bron, and Sébastien Anguille. "Chimeric Antigen Receptor-T-Cell Therapy for B-Cell Hematological Malignancies: An Update of the Pivotal Clinical Trial Data." Pharmaceutics 12, no. 2 (February 24, 2020): 194. http://dx.doi.org/10.3390/pharmaceutics12020194.

Full text
Abstract:
Chimeric antigen receptor (CAR)-T-cell therapy is an innovative form of adoptive cell therapy that has revolutionized the treatment of certain hematological malignancies, including B-cell non-Hodgkin lymphoma (NHL) and B-cell acute lymphoblastic leukemia (ALL). The treatment is currently also being studied in other B-cell neoplasms, including multiple myeloma (MM) and chronic lymphocytic leukemia (CLL). CD19 and B-cell maturation antigen (BCMA) have been the most popular target antigens for CAR-T-cell immunotherapy of these malignancies. This review will discuss the efficacy and toxicity data from the pivotal clinical studies of CD19- and BCMA-targeted CAR-T-cell therapies in relapsed/refractory B-cell malignancies (NHL, ALL, CLL) and MM, respectively.
APA, Harvard, Vancouver, ISO, and other styles
23

Sultana, Tanvira Afroze, Md Abdul Mottalib, Md Sirazul Islam, Mohiuddin Ahmed Khan, and Subhagata Choudhury. "rt-PCR method for diagnosis and follow-up of hematological malignancies: First approach in Bangladesh." Bangladesh Medical Research Council Bulletin 34, no. 1 (September 16, 2008): 1–11. http://dx.doi.org/10.3329/bmrcb.v34i1.1162.

Full text
Abstract:
Nested reverse-transcriptase polymerase chain reaction (rt-PCR) was performed on 58 leukemia patients at BIRDEM Laboratory, as a pioneering work in Bangladesh. Thirty of them were examined for the presence of BCR-ABL being clinically and morphologically diagnosed as chronic myeloid leukemia (CML) and 28 for PML-RARα fusion transcripts being clinically and morphologically diagnosed as acute promyelocytic leukemia (APL/ AML M3). The cases were selected for targeted therapy with imatinib mesylate and all-Trans retinoic acid (ATRA) to treat CML and APL respectively. Samples were received either before commencement or during therapy. In the positive cases, amplified DNA products were visible after gel electrophoresis and were reported accordingly. In case of BCR-ABL, positive results were found for five out of six (83.33%) untreated cases and 11 out of 24 (45.83%) treated cases. Positive results for PML-RARα were found for 12 out of 14 (85.70%) untreated cases and 11 out of 16 (68.75%) treated cases. A strong positive correlation was found between duration of treatment and negativity of PCR results in both the cases. In present times, the detection of minimal residual disease in patients undergoing treatment for hematological malignancies has become an important goal, not only to monitor the effectiveness of therapy but also to detect an impending relapse. This is the first time in Bangladesh that rt-PCR method is being employed to detect or monitor the presence of abnormal fusion genes in hematological malignancies. Keywords: Bangladesh; malignancy; rt-pcrDOI: 10.3329/bmrcb.v34i1.1162Bangladesh Med Res Counc Bull 2008; 34: 1-11
APA, Harvard, Vancouver, ISO, and other styles
24

Jose, Arun, Hind Rafei, and Jalil Ahari. "Combination targeted pulmonary hypertension therapy in the resolution of Dasatinib-associated pulmonary arterial hypertension." Pulmonary Circulation 7, no. 4 (July 5, 2017): 803–7. http://dx.doi.org/10.1177/2045893217716659.

Full text
Abstract:
Dasatinib is a small-molecule tyrosine kinase inhibitor used in the treatment of hematological malignancies. Pulmonary arterial hypertension (PAH) is a rare but known complication. The mainstay of treatment is cessation of Dasatinib, and while clinical improvement is rapid, complete hemodynamic resolution of pulmonary hypertension (PH) still remains exceedingly uncommon. We present a case of Dasatinib-induced PAH in a woman with chronic myeloid leukemia, who demonstrated rapid and complete clinical and hemodynamic resolution following treatment with combination pulmonary vasodilator therapy using an endothelin receptor antagonist and a phosphodiesterase-5 inhibitor. This case suggests there may be an association between the use of targeted PH medication in combination and the complete resolution of dasatinib-associated PAH, but further investigation is required.
APA, Harvard, Vancouver, ISO, and other styles
25

Kirchhoff, Hanna, Melanie Ricke-Hoch, Katharina Wohlan, Stefan Pietzsch, Ümran Karsli, Sergej Erschow, Robert Zweigerdt, et al. "Chemotherapy-Free Targeted Anti-BCR-ABL+ Acute Lymphoblastic Leukemia Therapy May Benefit the Heart." Cancers 14, no. 4 (February 15, 2022): 983. http://dx.doi.org/10.3390/cancers14040983.

Full text
Abstract:
Targeted therapies are currently considered the best cost–benefit anti-cancer treatment. In hematological malignancies, however, relapse rates and non-hematopoietic side effects including cardiotoxicity remain high. Here, we describe significant heart damage due to advanced acute lymphoblastic leukemia (ALL) with t(9;22) encoding the bcr-abl oncogene (BCR-ABL+ ALL) in murine xenotransplantation models. Echocardiography reveals severe cardiac dysfunction with impaired left ventricular function and reduced heart and cardiomyocyte dimensions associated with increased apoptosis. This cardiac damage is fully reversible, but cardiac recovery depends on the therapy used to induce ALL remission. Chemotherapy-free combination therapy with dasatinib (DAS), venetoclax (VEN) (targeting the BCR-ABL oncoprotein and mitochondrial B-cell CLL/Lymphoma 2 (BCL2), respectively), and dexamethasone (DEX) can fully revert cardiac defects, whereas the depletion of otherwise identical ALL in a genetic model using herpes simplex virus type 1 thymidine kinase (HSV-TK) cannot. Mechanistically, dexamethasone induces a pro-apoptotic BCL2-interacting mediator of cell death (BIM) expression and apoptosis in ALL cells but enhances pro-survival B-cell lymphoma extra-large (BCLXL) expression in cardiomyocytes and clinical recovery with the reversion of cardiac atrophy. These data demonstrate that therapies designed to optimize apoptosis induction in ALL may circumvent cardiac on-target side effects and may even activate cardiac recovery. In the future, combining the careful clinical monitoring of cardiotoxicity in leukemic patients with the further characterization of organ-specific side effects and signaling pathways activated by malignancy and/or anti-tumor therapies seems reasonable.
APA, Harvard, Vancouver, ISO, and other styles
26

Rahman, MM, and R. Rahim. "Flow Cytometric Immunophenotyping of Acute Leukemia the Essential Considerations." Pulse 9, no. 1 (March 14, 2017): 27–36. http://dx.doi.org/10.3329/pulse.v9i1.31878.

Full text
Abstract:
Objective: This article serves to promote the interest of participants involved in management of hematological malignancies in the efficacy of flow cytometry in the immunophenotypic characterization of leukemias and lymphomas. This well-defmed characterization plays a crucial role in diagnosis, classification, prognostic evaluation, and detection of minimal residual disease, in the context of clinical features and morphological diagnosis.Methodology: Relevant literature was retrieved to highlight the principles of operation of flow cytometly, applications, derivable clinical information, sources of errors, necessary antibodies and general considerations towards definitive diagnosis of acute leukemia.Conclusion: Hematological malignancy is an extremely heterogeneous disorder. Use of flow cytometric immunophenotyping in these disorders is already proven over two decades and allowed clinicians for proper classification along the latest WHO classification guidelines and will pave the way for targeted therapy and better patient management. The flow cytometric immunophenotyping services in the country will significantly outweigh the cost by reducing the duration of hospital stay and patient management in right way.Pulse Vol.9 January-December 2016 p.27-36
APA, Harvard, Vancouver, ISO, and other styles
27

de la Puente, Pilar, Barbara Muz, Feda Azab, Micah Luderer, and Abdel Kareem Azab. "Molecularly Targeted Therapies in Multiple Myeloma." Leukemia Research and Treatment 2014 (April 16, 2014): 1–8. http://dx.doi.org/10.1155/2014/976567.

Full text
Abstract:
Multiple myeloma (MM) is a hematological malignancy that remains incurable because most patients will eventually relapse or become refractory to the treatments. Although the treatments have improved, the major problem in MM is the resistance to therapy. Novel agents are currently in development for the treatment of relapsed/refractory MM, including immunomodulatory drugs, proteasome inhibitors, monoclonal antibodies, cell signaling targeted therapies, and strategies targeting the tumor microenvironment. We have previously reviewed in detail the contemporary immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies therapies for MM. Therefore, in this review, we focused on the role of molecular targeted therapies in the treatment of relapsed/refractory multiple myeloma, including cell signaling targeted therapies (HDAC, PI3K/AKT/mTOR, p38 MAPK, Hsp90, Wnt, Notch, Hedgehog, and cell cycle) and strategies targeting the tumor microenvironment (hypoxia, angiogenesis, integrins, CD44, CXCR4, and selectins). Although these novel agents have improved the therapeutic outcomes for MM patients, further development of new therapeutic agents is warranted.
APA, Harvard, Vancouver, ISO, and other styles
28

Almåsbak, Hilde, Tanja Aarvak, and Mohan C. Vemuri. "CAR T Cell Therapy: A Game Changer in Cancer Treatment." Journal of Immunology Research 2016 (2016): 1–10. http://dx.doi.org/10.1155/2016/5474602.

Full text
Abstract:
The development of novel targeted therapies with acceptable safety profiles is critical to successful cancer outcomes with better survival rates. Immunotherapy offers promising opportunities with the potential to induce sustained remissions in patients with refractory disease. Recent dramatic clinical responses in trials with gene modified T cells expressing chimeric antigen receptors (CARs) in B-cell malignancies have generated great enthusiasm. This therapy might pave the way for a potential paradigm shift in the way we treat refractory or relapsed cancers. CARs are genetically engineered receptors that combine the specific binding domains from a tumor targeting antibody with T cell signaling domains to allow specifically targeted antibody redirected T cell activation. Despite current successes in hematological cancers, we are only in the beginning of exploring the powerful potential of CAR redirected T cells in the control and elimination of resistant, metastatic, or recurrent nonhematological cancers. This review discusses the application of the CAR T cell therapy, its challenges, and strategies for successful clinical and commercial translation.
APA, Harvard, Vancouver, ISO, and other styles
29

Simpson-Haidaris, P. J., S. J. Pollock, S. Ramon, N. Guo, C. F. Woeller, S. E. Feldon, and R. P. Phipps. "Anticancer Role of PPARγAgonists in Hematological Malignancies Found in the Vasculature, Marrow, and Eyes." PPAR Research 2010 (2010): 1–36. http://dx.doi.org/10.1155/2010/814609.

Full text
Abstract:
The use of targeted cancer therapies in combination with conventional chemotherapeutic agents and/or radiation treatment has increased overall survival of cancer patients. However, longer survival is accompanied by increased incidence of comorbidities due, in part, to drug side effects and toxicities. It is well accepted that inflammation and tumorigenesis are linked. Because peroxisome proliferator-activated receptor (PPAR)-γagonists are potent mediators of anti-inflammatory responses, it was a logical extension to examine the role of PPARγagonists in the treatment and prevention of cancer. This paper has two objectives: first to highlight the potential uses for PPARγagonists in anticancer therapy with special emphasis on their role when used as adjuvant or combined therapy in the treatment of hematological malignancies found in the vasculature, marrow, and eyes, and second, to review the potential role PPARγand/or its ligands may have in modulating cancer-associated angiogenesis and tumor-stromal microenvironment crosstalk in bone marrow.
APA, Harvard, Vancouver, ISO, and other styles
30

Zhang, Chi, Tao Liu, Peng Luo, Li Gao, Xingyun Liao, Le Ma, Zhongyong Jiang, et al. "Near-infrared oxidative phosphorylation inhibitor integrates acute myeloid leukemia–targeted imaging and therapy." Science Advances 7, no. 1 (January 2021): eabb6104. http://dx.doi.org/10.1126/sciadv.abb6104.

Full text
Abstract:
Acute myeloid leukemia (AML) is a deadly hematological malignancy with frequent disease relapse. The biggest challenge for AML therapy is the lack of methods to target and kill the heterogeneous leukemia cells, which lead to disease relapse. Here, we describe a near-infrared (NIR) fluorescent dye, IR-26, which preferentially accumulates in the mitochondria of AML cells, depending on the hyperactive glycolysis of malignant cell, and simultaneously impairs oxidative phosphorylation (OXPHOS) to exert targeted therapeutic effects for AML cells. In particular, IR-26 also exhibits potential for real-time monitoring of AML cells with an in vivo flow cytometry (IVFC) system. Therefore, IR-26 represents a novel all-in-one agent for the integration of AML targeting, detection, and therapy, which may help to monitor disease progression and treatment responses, prevent unnecessary delays in administering upfront therapy, and improve therapeutic efficiency to the residual AML cells, which are responsible for disease relapse.
APA, Harvard, Vancouver, ISO, and other styles
31

Cronk, Robert J., Joanna Zurko, and Nirav N. Shah. "Bispecific Chimeric Antigen Receptor T Cell Therapy for B Cell Malignancies and Multiple Myeloma." Cancers 12, no. 9 (September 5, 2020): 2523. http://dx.doi.org/10.3390/cancers12092523.

Full text
Abstract:
Chimeric antigen receptor (CAR) modified T cell therapy offers a targeted immunotherapeutic approach to patients with refractory hematological malignancies. This technology is most advanced in B cell malignancies and multiple myeloma and is rapidly evolving as more data become available regarding clinical efficacy and response durability. Despite excellent initial response rates with single antigen targeting CARs, failure to respond to therapy and relapse due to target antigen downregulation remain clinical challenges. To mitigate immunophenotypic selective pressures, simultaneous dual antigen targeting with bispecific CAR T cells or multiple administration of different populations of CAR T cells may prevent relapse by addressing one resistance mechanism attributed to antigenic loss. This article will review recently published data on the use of dual targeting with CAR T cells from early phase clinical trials aimed at treating B cell malignancies and multiple myeloma.
APA, Harvard, Vancouver, ISO, and other styles
32

Zeidan, Amer M., Tehseen Salimi, and Robert S. Epstein. "Real-world use and outcomes of hypomethylating agent therapy in higher-risk myelodysplastic syndromes: why are we not achieving the promise of clinical trials?" Future Oncology 17, no. 36 (December 2021): 5163–75. http://dx.doi.org/10.2217/fon-2021-0936.

Full text
Abstract:
Myelodysplastic syndromes are hematological malignancies characterized by ineffective hematopoiesis and a high risk of progression to acute myeloid leukemia. Hypomethylating agents (HMAs), azacitidine and decitabine, are standard of care therapy for higher-risk myelodysplastic syndromes. However, outcomes reported for real-world studies fall short of those achieved in clinical trials. We conducted a targeted literature review exploring real-world utilization, persistence and outcomes with intravenous and subcutaneous HMA therapies to better understand barriers to achieving optimal outcomes in clinical practice. The potential benefits of oral HMA therapy were also explored. Underutilization and poor persistence with HMA therapy are associated with suboptimal outcomes, highlighting the need for approaches to improve utilization and persistence, so that patients achieve the optimum benefit from HMA therapy.
APA, Harvard, Vancouver, ISO, and other styles
33

Papanota, Aristea-Maria, Paraskevi Karousi, Christos K. Kontos, Ioannis Ntanasis-Stathopoulos, Andreas Scorilas, and Evangelos Terpos. "Multiple Myeloma Bone Disease: Implication of MicroRNAs in Its Molecular Background." International Journal of Molecular Sciences 22, no. 5 (February 27, 2021): 2375. http://dx.doi.org/10.3390/ijms22052375.

Full text
Abstract:
Multiple myeloma (MM) is a common hematological malignancy arising from terminally differentiated plasma cells. In the majority of cases, symptomatic disease is characterized by the presence of bone disease. Multiple myeloma bone disease (MMBD) is a result of an imbalance in the bone-remodeling process that leads to increased osteoclast activity and decreased osteoblast activity. The molecular background of MMBD appears intriguingly complex, as several signaling pathways and cell-to-cell interactions are implicated in the pathophysiology of MMBD. MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate the expression of their target mRNAs. Numerous miRNAs have been witnessed to be involved in cancer and hematological malignancies and their role has been characterized either as oncogenic or oncosuppressive. Recently, scientific research turned towards miRNAs as regulators of MMBD. Scientific data support that miRNAs finely regulate the majority of the signaling pathways implicated in MMBD. In this review, we provide concise information regarding the molecular pathways with a significant role in MMBD and the miRNAs implicated in their regulation. Moreover, we discuss their utility as molecular biomarkers and highlight the putative usage of miRNAs as novel molecular targets for targeted therapy in MMBD.
APA, Harvard, Vancouver, ISO, and other styles
34

Verma, Sheetal, Tamisha Vaughan, and Kevin D. Bunting. "Gab Adapter Proteins as Therapeutic Targets for Hematologic Disease." Advances in Hematology 2012 (2012): 1–10. http://dx.doi.org/10.1155/2012/380635.

Full text
Abstract:
The Grb-2 associated binder (Gab) family of scaffolding/adaptor/docking proteins is a group of three molecules with significant roles in cytokine receptor signaling. Gabs possess structural motifs for phosphorylation-dependent receptor recruitment, Grb2 binding, and activation of downstream signaling pathways through p85 and SHP-2. In addition, Gabs participate in hematopoiesis and regulation of immune response which can be aberrantly activated in cancer and inflammation. The multifunctionality of Gab adapters might suggest that they would be too difficult to consider as candidates for “targeted” therapy. However, the one drug/one target approach is giving way to the concept of one drug/multiple target approach since few cancers are addicted to a single signaling molecule for survival and combination drug therapies can be problematic. In this paper, we cover recent findings on Gab multi-functionality, binding partners, and their role in hematological malignancy and examine the concept of Gab-targeted therapy.
APA, Harvard, Vancouver, ISO, and other styles
35

Koudijs, Marco J., Lennart A. Kester, Jayne Y. Hehir-Kwa, Eugene T. P. Verwiel, Erik Strengman, Marc van Tuil, Douwe van der Leest, et al. "RNA-Sequencing Improves Diagnostics and Treatment of Pediatric Hematological Malignancies." Blood 138, Supplement 1 (November 5, 2021): 107. http://dx.doi.org/10.1182/blood-2021-147692.

Full text
Abstract:
Abstract Background Diagnosis and treatment of hematological malignancies relies increasingly on the detection of underlying genetic abnormalities. Various laboratory techniques, including karyotyping, SNP-array, FISH, MLPA and RT-PCR are typically required to detect the full spectrum of clinically relevant genetic aberrations. These techniques are also hampered in their sensitivity by their targeted approach or lack of resolution. Ideally, an unbiased genome wide approach like RNA sequencing (RNA-seq) as a one-test-fits-all, could save costs and efforts and streamline diagnostic procedures. In the Netherlands, the care for all children with oncological disorders has been concentrated in a single, national center. Within the Laboratory of Childhood Cancer Pathology, we aim for a comprehensive diagnostic pipeline by implementing RNA-seq to aid diagnosis, prognosis and treatment of all children with cancer in the Netherlands. Methods We have established an RNA-seq based diagnostic pipeline, primarily aimed at detecting gene fusion events. Library prep is performed on 50-300 ng total RNA isolated from fresh (frozen) samples, followed by ribo-depletion and subsequent paired-end sequencing (2x150 nt) using the Illumina NovaSeq platform. Data is analyzed using the StarFusion algorithm for gene-fusion detection. We are prospectively comparing the results with routine diagnostic procedures. In addition, we are validating the detection of single nucleotide variants (SNVs) from RNA-seq data and developing a diagnostic classifier, using a nearest neighbor network approach. Results Based on RNA-seq profiling in diagnostics for all patients entering the Princess Maxima Center, there are several use-cases that highlight the value of RNA-seq. 1) In a prospective cohort of 244 patients (pan-cancer, including 97 hematological malignancies) we have shown that the diagnostic yield for detecting gene fusion events increased by approximately 40% compared to classical methods. An example is the TNIP1--PDGFRB gene fusion in a patient with pre B-ALL, making this patient eligible for imatinib treatment, which was not detected by other methods. 2) Variant calling on RNA-seq shows that activating mutations in e.g. KRAS are detected with high sensitivity, stratifying patients for therapeutic MEK intervention. 3) By expression outlier analysis, we were able to detect various promotor exchanges, e.g. IGH-MYC or IGH--DUX4, which are typically hard to detect by molecular techniques since the genomic breakpoint is highly variable and no chimeric transcript is formed. 4) Preliminary results from our diagnostic classifier show its potential to predict subclasses of hematological malignancies, e.g. high-hyperdiploid or bi-phenotypic ALL patients. 5) Fusion gene breakpoints detected by RNA-seq serve as a target for MRD analysis, allowing us to monitor disease progression and therapy response in individual patients. Currently, RNA-seq data is available for more than 1500 pediatric tumor samples. At the upcoming conference we will present an update of our results and some typical cases highlighting the added value of RNA-seq in routine diagnostics. Conclusion We show that RNA-seq on pediatric cancer samples is feasible and of great value for routine diagnostics. It has a higher sensitivity to detect gene fusion events compared to targeted assays. RNA-seq based gene fusion detection, in combination with mutation and expression analysis, is also promising to improve classification of malignancies, prognosis and stratification of patients for targeted therapies. Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
36

Parker, Andy. "STP938, a Novel, Potent and Selective Inhibitor of CTP Synthase 1 (CTPS1) Is a Targeted Therapy Specifically Blocking De Novo Nucleotide Synthesis in Lymphomas and Leukemias." Blood 136, Supplement 1 (November 5, 2020): 31. http://dx.doi.org/10.1182/blood-2020-138436.

Full text
Abstract:
Introduction: The ability to reprogram cellular metabolism including enhanced de novo nucleotide synthesis is a hallmark of cancer and essential for tumor progression and survival. The pyrimidine synthesis pathway has been proposed as a metabolic sensitivity for cancer and over the past few decades several inhibitors of enzymes involved in the pathway have been evaluated in clinical trials (e.g. avicin, CPEC, PALA, leflunomide) primarily in solid tumors with limited efficacy and unacceptable toxicity. The final step in the pyrimidine de novo nucleotide synthesis pathway is the conversion of UTP to CTP. This step is unusual in the pathway in having two enzymes which catalyze the same reaction, CTP synthase 1 (CTPS1) and CTPS2, which have the same activity but very different cell type dependencies. CTPS1 has been shown to have an essential and non-redundant role in lymphocyte proliferation with the identification of an ultra-rare human hypomorphic mutation resulting in an 85% reduction in CTPS1 activity which manifests as a profound effect upon lymphoid cells but with no effects on other cell types. This hypomorphic mutation prevents proliferation of T-cells and B-cells following activation, thus selective inhibition of CTPS1 represents a novel targeted approach to inhibit proliferation of lymphocytes in hematological malignancies without inhibiting CTP synthesis in other cell types which utilise CTPS2. We have identified and optimized orally bioavailable selective inhibitors of CTPS1 (exemplified by STP938) and hypothesized that STP938 would demonstrate anti-tumor effects on cells derived from hematological malignancies. Methods: The in vitro anti-tumor activity of selective CTPS1 inhibition was investigated in a 2D monolayer assay in a panel of 200 cancer cell lines. Cell viability was measured with the Cell Titer-Blue Cell Viability Assay after 4 days incubation. Human PBMCs were isolated and T-cells were activated with anti CD3/CD28 antibodies to assess effects of CTPS1 inhibition on intracellular nucleotide levels which were quantified using Liquid Chromatography Mass Spectrometry (LC-MS). Jurkat T-ALL cells and WI38 embryonic fibroblasts were used to assess apoptosis which was quantified using the Caspase-Glo 3/7 Assay. Murine xenograft studies were conducted in NOD.SCID mice implanted with T-ALL cell lines to assess anti-tumor effects of CTPS1 inhibitors. Results: The panel of cancer cell lines consisted of 56 derived from hematological cancers and 141 derived from solid tumors. STP938, a potent and selective inhibitor of CTPS1 specifically blocked cell proliferation of the hematological cancers. 43 (77%) of the hematological cell lines had an IC50 &lt;100nM whereas only 15% of the solid tumor derived cell lines were affected by the CTPS1 inhibitor. The T-cell derived cell lines were extremely sensitive with many IC50 &lt;10nM. Profiling of intracellular nucleotide levels using activated human T-cells exposed to increasing concentrations of STP938 demonstrated the selective depletion of CTP but not ATP or GTP (see Figure). Jurkat T-ALL cells and WI38 fibroblasts were evaluated for markers of apoptosis in the presence of increasing concentrations of STP938. The Jurkat T-ALL cells exhibited a dose dependent increase in caspase 3/7 levels indicative of apoptosis. The WI38-fibroblast cells were unaffected. Murine xenograft studies using NHL T lymphoma and leukemia cell lines were conducted with oral dosing of CTPS1 inhibitor starting at the time of tumor cell inoculation or once tumors were established. In all cases, in a dose-dependent manner, CTPS1 inhibitors reduced or ablated tumor growth meeting NCI criteria for efficacy. Conclusions: Our preliminary results indicate that inhibition of CTPS1 enables cell specific inhibition of de novo pyrimidine nucleotide synthesis with a particular sensitivity displayed by cell lines derived from hematological malignancies. This disruption of de novo nucleotide synthesis by CTPS1 inhibition selectively induces apoptosis in lymphocyte cells but not in other cell types that are able to rely upon CTPS2 for their CTP synthesis. Orally bioavailable small molecule inhibitors of CTPS1 exert an anti-tumor effect in vivo in murine xenograft models and thus inhibition of CTPS1 represents a novel targeted approach to treat hematological malignancies. Figure Disclosures Parker: Step Pharma: Current Employment.
APA, Harvard, Vancouver, ISO, and other styles
37

Ana Sami. "Next Generation Stem Cells and their Implications in Cancer Therapy." Journal of the Pakistan Medical Association 73, no. 2 (January 25, 2023): S98—S104. http://dx.doi.org/10.47391/jpma.akus-16.

Full text
Abstract:
Stem cells have been implicated for decades in the treatment of hematological malignancies. These cells when isolated from the bone marrow, adipose tissue, or foetal tissue are deemed as the first generation of stem cells. The turn of the century saw the discovery of the second generation of stem cells such as the human Embryonic Stem Cells (hESCs) and induced Pluripotent Stem Cells (iPSCs). Advances in gene editing technology, in the past decade, have stimulated the rise of next-generation stem cells. Recent studies exploit the tumour tropism, multi-lineage differentiation, and auto-renewal capability of stem cells, and combine it with molecular biology techniques, to create potent anti-cancer therapies. Stem cells have been modified to have low immunogenicity and are thus being used as ‘trojan horses’ for the targeted, intra-tumoral delivery of anti-cancer drugs. continued...
APA, Harvard, Vancouver, ISO, and other styles
38

Angotzi, Francesco, Andrea Visentin, Federico Scarmozzino, Alessandro Cellini, Roberta Bertorelle, Marco Pizzi, Gianni Binotto, Angelo Paolo Dei Tos, and Livio Trentin. "Primary Myelofibrosis Occurring during Targeted Therapy for Chronic Lymphocytic Leukemia: A Report of Two Cases." Current Oncology 29, no. 3 (February 27, 2022): 1455–60. http://dx.doi.org/10.3390/curroncol29030122.

Full text
Abstract:
The disease course of chronic lymphocytic leukemia (CLL) is frequently characterized by the occurrence of various complications, such as second primary cancer, which can impact patients’ prognoses. While therapies for CLL have evolved tremendously in the past decades, overlooking the possibility of rare neoplasms that arise along with CLL may hinder the benefit that these therapies grant to patients. Moreover, the ability of newer therapies to alter the landscape of these complications is still largely unknown. Primary myelofibrosis (PMF) is not commonly associated with CLL, with only a few cases reported in the literature, with little information regarding the clinico-biological features and the optimal management for these associated conditions. Here, we report two unusual cases of PMF that occurred a few months after the start of therapy for CLL with targeted agents (ibrutinib and venetoclax). Both cases represented a diagnostic and therapeutic challenge, underscoring the need for clinicians to remain vigilant about the possible co-occurrence of these two hematological malignancies, especially in the era of targeted therapy for CLL.
APA, Harvard, Vancouver, ISO, and other styles
39

Saucier, Emilie, Florence Rabian, Hélène Boutroux, Marion Strullu, Karine Morand, Florian Chevillon, Benoit Brethon, Nicolas Boissel, Guy Leverger, and André Baruchel. "Azacitidine in Pediatric Hematologic Myeloid Malignancies: A Retrospective Study." Blood 134, Supplement_1 (November 13, 2019): 5130. http://dx.doi.org/10.1182/blood-2019-131062.

Full text
Abstract:
Purpose: Hypomethylating agents are approved in adults with Acute Myeloid Leukemia (AML) or Myelodysplastic Syndrome (MDS). By contrast, data in pediatric hematologic malignancies are scarce. Herein, we report the off-label administration of Azacitidine (Aza) in a cohort of children, adolescents and young adults (AYA). Methods: Patients from Robert-Debré, Saint-Louis and Armand-Trousseau University Hospitals (Assistance Publique-Hôpitaux de Paris), 25 year-old or younger at initiation of treatment and who received Aza between 2009 and 2019 were included. Data on indication, efficacy and toxicity of the treatment were retrospectively collected. Results: As 3 patients (pts) were treated twice with Aza at different stages of their disease, 35 treatments in 32 patients were analyzed. Median age at diagnosis was 11.7 y (range 0.1-22.4). Diagnosis were: 16 Acute Myeloid Leukemias (AML), 6 Juvenile Myelomonocytic Leukemias (JMML), 7 Myelodysplastic Syndromes (MDS), 2 Mixed Phenotype Acute Leukemias (MPAL) and 1 Interdigitating Dendritic Cells Sarcoma. Aza was administrated after a median of 2 lines of treatment (range 0-6) and front line in 5 out of 32 pts (16%; MDS = 4, JMML = 1). Concomitant therapy was administrated in 15/35 treatments (43%) (Sorafenib = 6, Gemtuzumab Ozogamicin = 2, donor lymphocyte infusions = 2, other = 5). Fourteen out of 35 treatments (40%) were delivered in patients with a history of hematopoietic stem cell transplantation (HSCT). Aza was administrated either intravenously (n = 11) or subcutaneously (n = 24) at a dose of 75mg/m² for 7 (20/35, 57%) or 5 (6/35, 17%) consecutive days every 28 days, i.e. same daily dose as in adults; at reduced doses (≤ 50mg/m²) in 3/35 patients; at a dosing scheme varying from one cycle to another in 6/35 patients. Patients received a median of 3 cycles of Aza per treatment (range 1-31). A total of 137 cycles was delivered. Aza was well tolerated with only 5/137 cycles delayed due to hematological (n = 3) or non-hematological (n = 2) toxicity. Aza was discontinued due to toxicity in 2/137 cycles (1 rectal bleeding, 1 severe sepsis). Anemia was noted in 68/130 cycles (52%), thrombocytopenia in 68/128 cycles (53%), neutropenia in 84/127 cycles (66%) and febrile neutropenia in 25/131 cycles (19%). Two grade 5 side effects were observed: 1 patient died from cerebral hemorrhage in a context of anti-platelet poly-immunization, another from septic shock in a context of neutropenia. Most common non-hematological side effects included: nausea, vomiting, diarrhea, subcutaneous injections lesions (local inflammation or hematoma) and infections. Responses to Aza are detailed in the attached table. Best responses to Aza included: 2 complete remissions (CR) both after 3 cycles, 3 CR with incomplete hematologic recovery (CRi) after a median of 2 cycles (range 1-2), 2 partial remissions (PR) after 2 and 3 cycles, 6 stable diseases (SD) during a median of 2 cycles (range 2-12) and 2 hematologic improvements. Overall response rate (sum of CR, CRi, PRs), was 19% (6/32) (3 non evaluable responses). Three out of the 6 patients treated with Aza combined with Sorafenib (AML = 4, MPAL = 2) obtained a response (CR: 2, CRi: 1). The association led to HSCT for 2 of them; the third patient had a CR maintained almost 3 years without HSCT, before relapsing on therapy. One patient with MDS had a SD for 12 months after palliative treatment with Aza. Overall, Aza treatment allowed to proceed to HSCT in 8 out of 32 patients (25%), 7 out of 8 proceeding to a first HSCT [CR: 1 patient, CRi: 3 patients, PR: 2 patients, SD: 1 patient, progressive disease: 1 patient]. These 8 pts included: 4 pts with AML, alive in CR after a median follow-up of 29.5 months (2 of them received concomitant targeted therapy); 2 pts with JMML (1 alive in CR after a follow-up of 25 months); 2 pts with MDS (both died, 7 and 30 months after Aza initiation). Finally, 24 out of 32 patients died and overall survival at 5 years was 23% (IC95% [11;48]) with no difference according to diagnosis. Conclusion: In a cohort of pediatric and AYA patients with heavily pretreated myeloid malignancies, Aza was well tolerated, and allowed to bridge 25% of patients to HSCT. Further prospective studies are needed to explore combination of Aza with targeted therapy in pediatric patients. Disclosures Boissel: NOVARTIS: Consultancy.
APA, Harvard, Vancouver, ISO, and other styles
40

Alrhmoun, Saleh, and Sergey Sennikov. "The Role of Tumor-Associated Antigen HER2/neu in Tumor Development and the Different Approaches for Using It in Treatment: Many Choices and Future Directions." Cancers 14, no. 24 (December 14, 2022): 6173. http://dx.doi.org/10.3390/cancers14246173.

Full text
Abstract:
The treatment of HER2-positive cancers has changed significantly over the past ten years thanks to a significant number of promising new approaches that have been added to our arsenal in the fight against cancer, including monoclonal antibodies, inhibitors of tyrosine kinase, antibody–drug conjugates, vaccination, and particularly, adoptive-T-cell therapy after its great success in hematological malignancies. Equally important is the new methodology for determining patients eligible for targeted HER2 therapy, which has doubled the number of patients who can benefit from these treatments. However, despite the initial enthusiasm, there are still several problems in this field represented by drug resistance and tumor recurrence that require the further development of new more efficient drugs. In this review, we discuss various approaches for targeting the HER2 molecule in cancer treatment, highlighting their benefits and drawbacks, along with the different mechanisms responsible for resistance to HER2-targeted therapies and how to overcome them.
APA, Harvard, Vancouver, ISO, and other styles
41

Gasic, Vladimir, Teodora Karan-Djurasevic, Djordje Pavlovic, Branka Zukic, Sonja Pavlovic, and Natasa Tosic. "Diagnostic and Therapeutic Implications of Long Non-Coding RNAs in Leukemia." Life 12, no. 11 (November 2, 2022): 1770. http://dx.doi.org/10.3390/life12111770.

Full text
Abstract:
Leukemia is a heterogenous group of hematological malignancies categorized in four main types (acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myeloid leukemia (CML) and chronic lymphocytic leukemia (CLL). Several cytogenetic and molecular markers have become a part of routine analysis for leukemia patients. These markers have been used in diagnosis, risk-stratification and targeted therapy application. Recent studies have indicated that numerous regulatory RNAs, such as long non-coding RNAs (lncRNAs), have a role in tumor initiation and progression. When it comes to leukemia, data for lncRNA involvement in its etiology, progression, diagnosis, treatment and prognosis is limited. The aim of this review is to summarize research data on lncRNAs in different types of leukemia, on their expression pattern, their role in leukemic transformation and disease progression. The usefulness of this information in the clinical setting, i.e., for diagnostic and prognostic purposes, will be emphasized. Finally, how particular lncRNAs could be used as potential targets for the application of targeted therapy will be considered.
APA, Harvard, Vancouver, ISO, and other styles
42

Moore, Joseph O. "Tyrosine Kinase Inhibitors for Chronic Myeloid Leukemia—Is There an Optimal Treatment Plan?" Oncology & Hematology Review (US) 02 (2009): 34. http://dx.doi.org/10.17925/ohr.2009.02.0.34.

Full text
Abstract:
Chronic myeloid leukemia (CML) is the first hematological malignancy in which understanding of the disease pathogenesis has led to rational design therapy. Imatinib mesylate, a first-generation tyrosine kinase inhibitor (TKI), is the current standard of care for patients with chronic-phase CML (CML-CP). It is the only targeted therapy in CML with proven long-term efficacy and safety. Two potent second-generation TKIs, nilotinib and dasatinib, are currently approved for patients who are resistant to or intolerant of imatinib or who are diagnosed with advanced disease. Alternatives for front-line TKI therapy are under active investigation and emerging data indicate that first-line therapy with high-dose imatinib, dasatinib, or nilotinib may be of benefit. Selection of a second-line therapy should consider the side-effect profile, patient comorbidities, type and stage of disease, mutational status, and other practical considerations.
APA, Harvard, Vancouver, ISO, and other styles
43

Thirukkumaran, Chandini M., and Don G. Morris. "Oncolytic Virotherapy for Multiple Myeloma: Past, Present, and Future." Bone Marrow Research 2011 (May 10, 2011): 1–10. http://dx.doi.org/10.1155/2011/632948.

Full text
Abstract:
Multiple myeloma (MM) is a B-cell malignancy that is currently felt to be incurable. Despite recently approved novel targeted treatments such as lenalidomide and bortezomib, most MM patients' relapse is emphasizing the need for effective and well-tolerated therapies for this deadly disease. The use of oncolytic viruses has garnered significant interest as cancer therapeutics in recent years, and are currently under intense clinical investigation. Both naturally occurring and engineered DNA and RNA viruses have been investigated preclinically as treatment modalities for several solid and hematological malignancies. Presently, only a genetically modified measles virus is in human clinical trials for MM. The information obtained from this and other future clinical trials will guide clinical application of oncolytic viruses as anticancer agents for MM. This paper provides a timely overview of the history of oncolytic viruses for the treatment of MM and future strategies for the optimization of viral therapy for this disease.
APA, Harvard, Vancouver, ISO, and other styles
44

Ravi, Dashnamoorthy, Afshin Beheshti, Kristine Burgess, Athena Kritharis, Ying Chen, Andrew M. Evens, and Biju Parekkadan. "An Analysis of Transcriptomic Burden Identifies Biological Progression Roadmaps for Hematological Malignancies and Solid Tumors." Biomedicines 10, no. 11 (October 27, 2022): 2720. http://dx.doi.org/10.3390/biomedicines10112720.

Full text
Abstract:
Biological paths of tumor progression are difficult to predict without time-series data. Using median shift and abacus transformation in the analysis of RNA sequencing data sets, natural patient stratifications were found based on their transcriptomic burden (TcB). Using gene-behavior analysis, TcB groups were evaluated further to discover biological courses of tumor progression. We found that solid tumors and hematological malignancies (n = 4179) share conserved biological patterns, and biological network complexity decreases at increasing TcB levels. An analysis of gene expression datasets including pediatric leukemia patients revealed TcB patterns with biological directionality and survival implications. A prospective interventional study with PI3K targeted therapy in canine lymphomas proved that directional biological responses are dynamic. To conclude, TcB-enriched biological mechanisms detected the existence of biological trajectories within tumors. Using this prognostic informative novel informatics method, which can be applied to tumor transcriptomes and progressive diseases inspires the design of progression-specific therapeutic approaches.
APA, Harvard, Vancouver, ISO, and other styles
45

Yang, Fan, Zhihui Li, Changwei Dong, Yang LEI, YanPing Geng, XiaoPei Wen, Yan Zhou, and Tong Wu. "Daratumumab and Venetoclax-Containing Regimens in the Management of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation in Hematological Malignancies." Blood 136, Supplement 1 (November 5, 2020): 9. http://dx.doi.org/10.1182/blood-2020-141766.

Full text
Abstract:
Background: The prognosis of relapse after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with hematological malignancies is poor with conventional therapies such as immunosuppressant withdrawal, interferon, chemotherapy, donor lymphocyte infusion and second allo-HSCT. Immunotherapy and targeted therapy may bring new hope for this setting. Objective: In current study, the safety and efficacy of daratumumab and venetoclax-containing regimens in the management of relapse after allo-HSCT in hematological malignancies were evaluated. Methods: From August 2019 to May 2020, 12 patients with hematological malignancies who relapsed after allo-HSCT were included. The median age was 26 (5-49) years old. The diagnosis included acute myeloid leukemia (AML, n=3), acute lymphoblastic leukemia (B-ALL, n=2), T lymphoblastic lymphoma/leukemia (T-LBL, n=6) and B lymphoblastic lymphoma/leukemia (B-LBL, n=1). The disease status before transplant was complete remission (CR) in 8 cases (minimal residual disease (MRD)+ in 4, MRD- in 4), partial remission (PR) in 1 case and recurrence in 3 cases. The types of transplant were haploidentical in 6 cases (50%), unrelated in 5 cases (41.7%) and matched sibling in 1 case (8.3%). Myeloablative conditioning regimens were used and GVHD prophylaxis was with cyclosporine, short-term methotrexate and mycophenolate mofetil. Anti-thymocyte globulin was applied in haploidentical and unrelated transplants. The median relapse time was 4 (1-10) months after HSCT and the pattern of relapse included hematologic relapse in 11 cases (91.7%), and MRD+ in 1 case (8.3%). The expression of CD38 antigen was detected by flow cytometry. Daratumumab (400mg for children, 800mg for adults at day 0 and 14) and venetoclax (50mg/d, gradually increased to 100-400mg/d, d1-28) were administrated for induction and consolidation therapies. Results After 1 cycle of induction therapy, 5 cases achieved CR (41.7%) (4/6 T-LBL, 1/3 AML), 1 case in PR (T-LBL, 8.3%), and 6 cases in non-remission (NR) (2/2 B-ALL, 2/3 AML, 1/1 B-LBL, 1/6 T-LBL, 50%). The median time to achieve CR was 26 (18-42) days. Three of 5 CR patients were treated with daratumumab and venetoclax only, and rest 2 CR patients were treated with daratumumab and venetoclax plus cytarabine (100mg/d for 3-5d) and etoposide (100mg/d for 3-5d). Four patients in CR were consolidated with daratumumab and venetoclax for continuous MRD- CR with the median 263 days(210,335) , while the rest 1 patient in CR underwent the second allo-HSCT.The median consolidation therapies was 2 (1-4) cycles and the interval was 8 weeks. With the median follow-up 236 (138-365) days, eight of 12 patients (66.7%) survived and DFS was 41.7%. Seven patients developed treatment-related neutropenia (58.3%) and 4 patients had infections (33.3%). No severe side effects were noted. No GVHD occurred during treatment with above regimens. Conclusion: Our pilot clinical study has shown that daratumumab and venetoclax-containing regimens are safe and effective in the management of relapse after allo-HSCT in patients with hematological malignancies. It seems that better response are in T-LBL. It may provide an option for the patients their leukemic cells expressing CD38 with or without chemotherapy-resistant diseases. [Key words] relapse; allogeneic hematopoietic stem cell transplantation; hematological malignancy; daratumumab; venetoclax Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
46

Barros, Luciana Rodrigues Carvalho, Samuel Campanelli Freitas Couto, Daniela da Silva Santurio, Emanuelle Arantes Paixão, Fernanda Cardoso, Viviane Jennifer da Silva, Paulo Klinger, et al. "Systematic Review of Available CAR-T Cell Trials around the World." Cancers 14, no. 11 (May 27, 2022): 2667. http://dx.doi.org/10.3390/cancers14112667.

Full text
Abstract:
In this systematic review, we foresee what could be the approved scenario in the next few years for CAR-T cell therapies directed against hematological and solid tumor malignancies. China and the USA are the leading regions in numbers of clinical studies involving CAR-T. Hematological antigens CD19 and BCMA are the most targeted, followed by mesothelin, GPC3, CEA, MUC1, HER2, and EGFR for solid tumors. Most CAR constructs are second-generation, although third and fourth generations are being largely explored. Moreover, the benefit of combining CAR-T treatment with immune checkpoint inhibitors and other drugs is also being assessed. Data regarding product formulation and administration, such as cell phenotype, transfection technique, and cell dosage, are scarce and could not be retrieved. Better tracking of trials’ status and results on the ClinicalTrials.gov database should aid in a more concise and general view of the ongoing clinical trials involving CAR-T cell therapy.
APA, Harvard, Vancouver, ISO, and other styles
47

Akbari, Parvin, Elisabeth J. M. Huijbers, Maria Themeli, Arjan W. Griffioen, and Judy R. van Beijnum. "The tumor vasculature an attractive CAR T cell target in solid tumors." Angiogenesis 22, no. 4 (October 18, 2019): 473–75. http://dx.doi.org/10.1007/s10456-019-09687-9.

Full text
Abstract:
Abstract T cells armed with a chimeric antigen receptor, CAR T cells, have shown extraordinary activity against certain B lymphocyte malignancies, when targeted towards the CD19 B cell surface marker. These results have led to the regulatory approval of two CAR T cell approaches. Translation of this result to the solid tumor setting has been problematic until now. A number of differences between liquid and solid tumors are likely to cause this discrepancy. The main ones of these are undoubtedly the uncomplicated availability of the target cell within the blood compartment and the abundant expression of the target molecule on the cancerous cells in the case of hematological malignancies. Targets expressed by solid tumor cells are hard to engage due to the non-adhesive and abnormal vasculature, while conditions in the tumor microenvironment can be extremely immunosuppressive. Targets in the tumor vasculature are readily reachable by CAR T cells and reside outside the immunosuppressive tumor microenvironment. It is therefore hypothesized that targeting CAR T cells towards the tumor vasculature of solid tumors may share the excellent effects of CAR T cell therapy with that against hematological malignancies. A few reports have shown promising results. Suggestions are provided for further improvement.
APA, Harvard, Vancouver, ISO, and other styles
48

Sorrentino, Vincent G., Srijan Thota, Edward A. Gonzalez, Pranela Rameshwar, Victor T. Chang, and Jean-Pierre Etchegaray. "Hypomethylating Chemotherapeutic Agents as Therapy for Myelodysplastic Syndromes and Prevention of Acute Myeloid Leukemia." Pharmaceuticals 14, no. 7 (July 4, 2021): 641. http://dx.doi.org/10.3390/ph14070641.

Full text
Abstract:
Myelodysplastic Syndromes (MDSs) affect the elderly and can progress to Acute Myeloid Leukemia (AML). Epigenetic alterations including DNA methylation and chromatin modification may contribute to the initiation and progression of these malignancies. DNA hypomethylating agents such as decitabine and azacitidine are used as therapeutic treatments and have shown to promote expression of genes involved in tumor suppression, apoptosis, and immune response. Another anti-cancer drug, the proteasome inhibitor bortezomib, is used as a chemotherapeutic treatment for multiple myeloma (MM). Phase III clinical trials of decitabine and azacitidine used alone and in combination with other chemotherapeutics demonstrated their capacity to treat hematological malignancies and prolong the survival of MDS and AML patients. Although phase III clinical trials examining bortezomib’s role in MDS and AML patients are limited, its underlying mechanisms in MM highlight its potential as a chemotherapeutic for such malignancies. Further research is needed to better understand how the epigenetic mechanisms mediated by these chemotherapeutic agents and their targeted gene networks are associated with the development and progression of MDS into AML. This review discusses the mechanisms by which decitabine, azacitidine, and bortezomib alter epigenetic programs and their results from phase III clinical trials.
APA, Harvard, Vancouver, ISO, and other styles
49

Cheng, Wei, Tian-tian Yu, Ai-ping Tang, Ken He Young, and Li Yu. "Blastic Plasmacytoid Dendritic Cell Neoplasm: Progress in Cell Origin, Molecular Biology, Diagnostic Criteria and Therapeutic Approaches." Current Medical Science 41, no. 3 (June 2021): 405–19. http://dx.doi.org/10.1007/s11596-021-2393-3.

Full text
Abstract:
SummaryBlastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare hematological malignancy characterized by recurrent skin nodules, an aggressive clinical course with rapid involvement of hematological organs, and a poor prognosis with poor overall survival. BPDCN is derived from plasmacytoid dendritic cells (pDCs) and its pathogenesis is unclear. The tumor cells show aberrant expression of CD4, CD56, interleukin-3 receptor alpha chain (CD123), blood dendritic cell antigen 2 (BDCA 2/CD303), blood dendritic cell antigen 4 (BDCA4) and transcription factor (E protein) E2-2 (TCF4). The best treatment drugs are based on experience by adopting those used for either leukemia or lymphoma. Relapse with drug resistance generally occurs quickly. Stem cell transplantation after the first complete remission is recommended and tagraxofusp is the first targeted therapy. In this review, we summarize the differentiation of BPDCN from its cell origin, its connection with normal pDCs, clinical characteristics, genetic mutations and advances in treatment of BPDCN. This review provides insights into the mechanisms of and new therapeutic approaches for BPDCN.
APA, Harvard, Vancouver, ISO, and other styles
50

Tai, Waqqas, Ahsan Wahab, Diana Franco, Zunairah Shah, Aqsa Ashraf, Qurrat-Ul-Ain Abid, Yaqub Nadeem Mohammed, Darshan Lal, and Faiz Anwer. "Emerging Role of Antibody-Drug Conjugates and Bispecific Antibodies for the Treatment of Multiple Myeloma." Antibodies 11, no. 2 (March 24, 2022): 22. http://dx.doi.org/10.3390/antib11020022.

Full text
Abstract:
Multiple myeloma (MM) is characterized by malignant proliferation of malignant plasma cells; it is the second most common hematological malignancy associated with significant morbidity. Genetic intricacy, instability, and diverse clinical presentations remain a barrier to cure. The treatment of MM is modernized with the introduction of newer therapeutics agents, i.e., target-specific monoclonal antibodies. The currently available literature lacks the benefits of newer targeted therapy being developed with an aim to reduce side effects and increase effectiveness, compared to conventional chemotherapy regimens. This article aims to review literature about the current available monoclonal antibodies, antibody-drug conjugates, and bispecific antibodies for the treatment of MM.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography