90 results on '"Bogels M"'
Search Results
2. Is There a Role for Daratumumab Retreatment in Patients with Relapsed/Refractory Multiple Myeloma?
- Author
-
Galusic, Davor, Krecak, Ivan, Blaslov, Viktor, Krstulovic Opara, Andela, Valkovic, Toni, and Basic Kinda, Sandra
- Subjects
CHIMERIC antigen receptors ,BLOOD diseases ,MULTIPLE myeloma ,DARATUMUMAB ,PLASMA cells - Abstract
Multiple myeloma (MM) is a hematologic disease characterized by the clonal expansion of malignant plasma cells that accumulate in the bone marrow, leading to osteolytic bone disease, hypercalcemia, anemia, and renal dysfunction. Daratumumab was the first monoclonal anti-CD38 antibody approved for the treatment of MM, initially in relapse/refractory settings and, more recently, for newly diagnosed patients. Increased first-line usage of daratumumab will also substantially change treatment approaches for patients with relapsed/refractory disease. Due to the cost and availability of bispecific T cell redirecting antibodies (BsAbs) and chimeric antigen receptor T cell therapy (CAR-T) in real-life settings in many countries, retreatment with daratumumab in subsequent lines of therapy might be a reasonable choice. Data regarding efficacy and optimal combinations of daratumumab retreatment are lacking, and here we provide a short literature review of available data. We identified only a small number of articles based on retrospective analysis of medical records in real-life settings. A strong consistency in results regarding response rates and treatment duration was noticed among mainly heavily pre-treated MM patients, with approximately half of patients achieving at least partial remission (PR) after retreatment with daratumumab-based protocol. The duration of treatment and time to the next treatment for retreatment episodes were considerable and consistent with clinical expectations for later lines of therapy. The analysis of data in this literature review indicates that daratumumab retreatment may provide meaningful clinical benefit to some patients with relapsed/refractory MM despite having prior exposure. However, further research is needed to identify clinical and biological parameters that may predict favorable responses to daratumumab retreatment. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
3. Greater Omentum: Multifaceted Interactions in Neurological Recovery and Disease Progression.
- Author
-
Xiang Li, Yuchuan Ding, Haddad, Yazeed W., and Xiaokun Geng
- Subjects
DISEASE progression ,NEUROLOGICAL disorders - Abstract
The greater omentum, a unique anatomical structure composed of adipocytes, loose connective tissue, and a dense vascular network. Plays a pivotal role beyond its traditional understanding. It houses specialized immunological units known as 'Milky spots,' making it a key player in immune response. Moreover, the omentum's capacity to enhance tissue perfusion, absorb edema fluid, boost acetylcholine synthesis, and foster neuron repair have rendered it a topic of interest in the context of various diseases, especially neurological disorders. This review provides a comprehensive overview of the intricate anatomy and histology of the greater omentum, casting light on its multifaceted functions and its associations with a spectrum of diseases. With a specific focus on neurological ailments, we delineate the intricate relationship that the omentum shares with other pathologies like stroke and we underly its contribution to serving as a therapeutic agent in neurological disorders. By deciphering the underlying mechanisms and emphasizing areas that demand further investigation. This review aims to spark renewed interest and pave the way for comprehensive studies exploring the greater omentum's potential in neurology and broader medicine overall. Given these diverse interactions that yet remain elusive, we must investigate and understand the nuanced relationship between the greater omentum and pathologies, especially its role in stroke's pathophysiology and therapeutic interventions so as to enhance patient care. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Daratumumab in transplant-eligible patients with newly diagnosed multiple myeloma: final analysis of clinically relevant subgroups in GRIFFIN.
- Author
-
Chari, Ajai, Kaufman, Jonathan L., Laubach, Jacob, Sborov, Douglas W., Reeves, Brandi, Rodriguez, Cesar, Silbermann, Rebecca, Costa, Luciano J., Anderson Jr, Larry D., Nathwani, Nitya, Shah, Nina, Bumma, Naresh, Holstein, Sarah A., Costello, Caitlin, Jakubowiak, Andrzej, Wildes, Tanya M., Orlowski, Robert Z., Shain, Kenneth H., Cowan, Andrew J., and Pei, Huiling
- Subjects
MULTIPLE myeloma ,DARATUMUMAB ,PROGRESSION-free survival ,LENALIDOMIDE ,CONFIDENCE intervals - Abstract
The randomized, phase 2 GRIFFIN study (NCT02874742) evaluated daratumumab plus lenalidomide/bortezomib/dexamethasone (D-RVd) in transplant-eligible newly diagnosed multiple myeloma (NDMM). We present final post hoc analyses (median follow-up, 49.6 months) of clinically relevant subgroups, including patients with high-risk cytogenetic abnormalities (HRCAs) per revised definition (del[17p], t[4;14], t[14;16], t[14;20], and/or gain/amp[1q21]). Patients received 4 induction cycles (D-RVd/RVd), high-dose therapy/transplant, 2 consolidation cycles (D-RVd/RVd), and lenalidomide±daratumumab maintenance (≤ 2 years). Minimal residual disease–negativity (10
−5 ) rates were higher for D-RVd versus RVd in patients ≥ 65 years (67.9% vs 17.9%), with HRCAs (54.8% vs 32.4%), and with gain/amp(1q21) (61.8% vs 28.6%). D-RVd showed a trend toward improved progression-free survival versus RVd (hazard ratio [95% confidence interval]) in patients ≥ 65 years (0.29 [0.06–1.48]), with HRCAs (0.38 [0.14–1.01]), and with gain/amp(1q21) (0.42 [0.14–1.27]). In the functional high-risk subgroup (not MRD negative at the end of consolidation), the hazard ratio was 0.82 (0.35–1.89). Among patients ≥ 65 years, grade 3/4 treatment-emergent adverse event (TEAE) rates were higher for D-RVd versus RVd (88.9% vs 77.8%), as were TEAEs leading to discontinuation of ≥ 1 treatment component (37.0% vs 25.9%). One D-RVd patient died due to an unrelated TEAE. These results support the addition of daratumumab to RVd in transplant-eligible patients with high-risk NDMM. Video Abstract -YjyZJrsb3fs1sAVfK2kfK [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
5. Immune profiling of patients with extranodal natural killer/T cell lymphoma treated with daratumumab.
- Author
-
Qing, Min, Zhou, Tianyuan, Perova, Tatiana, Abraham, Yann, Sweeney, Cheryl, Krevvata, Maria, Zhang, Xiaokang, Qi, Ming, Gao, Grace, Kim, Tae Min, Yao, Ming, Cho, Seok-Goo, Eom, Hyeon Seok, Lim, Soon Thye, Yeh, Su-peng, Kwong, Yok Lam, Yoon, Dok Hyun, Kim, Jin Seok, Kim, Won Seog, and Zhou, Longen
- Subjects
T cells ,DARATUMUMAB ,NON-Hodgkin's lymphoma ,ANTINEOPLASTIC agents ,LYMPHOMAS - Abstract
Natural killer/T cell lymphoma (NKTCL) is a highly aggressive, heterogeneous non-Hodgkin lymphoma resulting from malignant proliferation of cytotoxic natural killer (NK) or T cells. Previous studies demonstrated variable expression of CD38 on NKTCL tumors. Daratumumab, a human IgGκ monoclonal antibody targeting CD38 with a direct on-tumor and immunomodulatory mechanism of action, was hypothesized to be a novel therapeutic option for patients with relapsed or refractory (R/R) NKTCL. In the phase 2 NKT2001 study (ClinicalTrials.gov Identifier: NCT02927925) assessing the safety and efficacy of daratumumab, a suboptimal overall response rate was seen in R/R NKTCL patients. One patient, whose tumors did not express CD38, responded to treatment, suggesting that the immunomodulatory activities of daratumumab may be sufficient to confer clinical benefit. To understand the suboptimal response rate and short duration of response, we investigated the immune profile of NKTCL patients from NKT2001 in the context of daratumumab anti-tumor activity. Tumor tissue and whole blood were, respectively, analyzed for CD38 expression and patient immune landscapes, which were assessed via cytometry by time-of-flight (CyTOF), multiparameter flow cytometry (MPFC), clonal sequencing, and plasma Epstein-Barr virus (EBV)-DNA level measurements. Changes observed in the immune profiles of NKTCL patients from NKT2001, including differences in B and T cell populations between responders and nonresponders, suggest that modulation of the immune environment is crucial for daratumumab anti-tumor activities in NKTCL. In conclusion, these findings highlight that the clinical benefit of daratumumab in NKTCL may be enriched by B/T cell–related biomarkers. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Subcutaneous daratumumab in Chinese patients with relapsed or refractory multiple myeloma: an open-label, multicenter, phase 1 study (MMY1010).
- Author
-
Gang An, Zheng Ge, Hongmei Jing, Jing Liu, Guoping Yang, Ru Feng, Zhongyuan Xu, Ming Qi, Jianping Wang, Juanjuan Song, Wei Zhou, Binbin Sun, Dian Zhu, Xi Chen, Canchan Cui, and Lugui Qiu
- Published
- 2024
- Full Text
- View/download PDF
7. Daratumumab plus lenalidomide/dexamethasone in untreated multiple myeloma: analysis of key subgroups of the MAIA study.
- Author
-
Moreau P, Facon T, Usmani SZ, Bahlis N, Raje N, Plesner T, Orlowski RZ, Basu S, Nahi H, Hulin C, Quach H, Goldschmidt H, O'Dwyer M, Perrot A, Venner CP, Weisel K, Tiab M, Macro M, Frenzel L, Leleu X, Wang G, Pei H, Krevvata M, Carson R, Borgsten F, and Kumar SK
- Abstract
In the MAIA study (median follow-up, 56.2 months), daratumumab plus lenalidomide and dexamethasone (D-Rd) significantly improved progression-free survival (PFS) and overall survival versus lenalidomide and dexamethasone (Rd) alone in transplant-ineligible newly diagnosed multiple myeloma (NDMM). In this post hoc analysis of clinically important subgroups in MAIA (median follow-up, 64.5 months), transplant-ineligible patients with NDMM were randomized 1:1 to D-Rd or Rd. The primary endpoint was PFS; secondary endpoints included overall response rate (ORR) and measurable residual disease (MRD)-negativity rate (10
-5 ). PFS favored D-Rd versus Rd in most subgroups, including patients aged ≥75 years (HR, 0.59; 95% CI, 0.44-0.79), frail patients (HR, 0.64; 95% CI, 0.48-0.85), patients with high-risk cytogenetics (HR, 0.59; 95% CI, 0.44-0.80), and patients with isolated gain(1q21) (HR, 0.36; 95% CI, 0.19-0.67). ORRs, MRD-negativity rates, and sustained (≥12 months) MRD-negativity rates were higher with D-Rd versus Rd across subgroups. In patients aged ≥75 years, rates of grade 3/4 and serious treatment-emergent adverse events (TEAEs) were similar for D-Rd and Rd, but discontinuation due to TEAEs was lower for D-Rd. Results support use of D-Rd for high-risk patients, supporting D-Rd as a standard of care for transplant-ineligible NDMM. This trial was registered at www.clinicaltrials.gov as NCT02252172., Competing Interests: Competing interests: PM served on an advisory board for and received honoraria from Janssen, Celgene, Amgen, AbbVie, Sanofi, and Oncopeptides. TF has nothing to disclose. SZU received research funding from Amgen, Array BioPharma, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Janssen, Merck, Pharmacyclics, Sanofi, Seattle Genetics, SkylineDx, and Takeda; served in a consulting role for AbbVie, Amgen, Bristol Myers Squibb, Celgene, Edo Pharma, Genentech, Gilead, GlaxoSmithKline, Janssen, Oncopeptides, Sanofi, Seattle Genetics, Secura Bio, SkylineDx, Takeda, and TeneoBio; and served on a speakers bureau for Amgen, Bristol Myers Squibb, Janssen, and Sanofi. NB served in a consulting or advisory role for and received honoraria from AbbVie, Bristol Myers Squibb/Celgene, FORUS, Genentech, Janssen, Karyopharm, Pfizer, Sanofi, and Takeda; and received research funding from Pfizer and Celgene. NR served as a consultant for Bristol Myers Squibb, Janssen, Takeda, Amgen, and GlaxoSmithKline; and received research funding from bluebird bio. TP served as an advisor for Janssen, Celgene, Takeda, Oncopeptides, Genentech, CSL Behring, and AbbVie; and received research support from Janssen, Genmab, Celgene, Takeda, Oncopeptides, Genentech, AbbVie, and Roche. RZO received research funding from Asylia Therapeutics, Biotheryx, Heidelberg Pharma, CARsgen, Celgene/Bristol Myers Squibb, Exelixis, Janssen, Sanofi-Aventis, and Takeda; received honoraria from and served as a member on a board of directors or advisory committee for AbbVie, Biotheryx, Bristol Myers Squibb, Janssen, Karyopharm, Meridian Therapeutics, Monte Rosa Therapeutics, Neoleukin, Oncopeptides, Regeneron, Sanofi-Aventis, and Takeda; and holds stock in Asylia Therapeutics. SB served as a consultant and on a speakers bureau for Sanofi, Pfizer, and Bristol Myers Squibb. HN is an employee of Genmab. CH received honoraria from Janssen, Bristol Myers Squibb, Amgen, Takeda, and AbbVie. HQ served as a consultant for and received research funding from AbbVie, Bristol Myers Squibb, Karyopharm, Amgen, GlaxoSmithKline, and Antengene. HG received grants and/or provisions of Investigational Medicinal Product from Amgen, Array BioPharma/Pfizer, Bristol Myers Squibb/Celgene, Chugai, Dietmar Hopp Foundation, Janssen, Johns Hopkins University, Mundipharma, and Sanofi; received research support from Amgen, Bristol Myers Squibb, Celgene, GlycoMimetics, GlaxoSmithKline, Heidelberg Pharma, Roche, Karyopharm, Janssen, Incyte, Millennium Pharmaceuticals, Molecular Partners, Merck Sharp & Dohme, MorphoSys AG, Pfizer, Sanofi, Takeda, and Novartis; served on an advisory board for Amgen, Bristol Myers Squibb, Janssen, Sanofi, and Adaptive Biotechnologies; received honoraria from Amgen, Bristol Myers Squibb, Chugai, GlaxoSmithKline, Janssen, Novartis, Sanofi, and Pfizer; and received support for attending meetings and/or travel from Amgen, Bristol Myers Squibb, GlaxoSmithKline, Janssen, Novartis, Sanofi, and Pfizer. MO served as a consultant for Janssen. AP received honoraria from AbbVie, Amgen, Celgene/Bristol Myers Squibb, GlaxoSmithKline, Janssen, Sanofi, and Takeda; served as a member on a board or advisory committee for Amgen, Celgene/Bristol Myers Squibb, GlaxoSmithKline, Janssen, Pfizer, and Takeda; and received research funding from Takeda. CPV received honoraria from Janssen, Bristol Myers Squibb, Sanofi, FORUS, Pfizer, AbbVie, GlaxoSmithKline, and Amgen. KW received research funding from Amgen, Adaptive Biotechnologies, AstraZeneca, Bristol Myers Squibb/Celgene, BeiGene, Janssen, GlaxoSmithKline, Sanofi, Stemline, and Takeda; received honoraria from AbbVie, Amgen, Adaptive Biotechnologies, AstraZeneca, Bristol Myers Squibb/Celgene, BeiGene, Janssen, GlaxoSmithKline, Karyopharm, Novartis, Oncopeptides, Pfizer, Roche, Sanofi, Stemline, and Takeda; and received travel support from Janssen, GlaxoSmithKline, and Sanofi. MT has nothing to disclose. MM served on an advisory board for Celgene/Bristol Myers Squibb, Janssen, Takeda, Amgen, GlaxoSmithKline, and Sanofi; received research funding from Janssen and Takeda; and received accommodations from Janssen, Celgene/Bristol Myers Squibb, Takeda, Amgen, and Sanofi. LF received honoraria from Janssen, Sanofi, Amgen, and Takeda; and received funding from Janssen and Amgen. XL received honoraria, research support, and consulting fees from Amgen, Merck, Bristol Myers Squibb, GlaxoSmithKline, Janssen, Oncopeptides, Takeda, Roche, Novartis, AbbVie, Sanofi, Gilead, Pfizer, Harpoon Therapeutics, Regeneron, and iTeos. GW, HP, and FB are employees of Janssen and hold stock in Johnson & Johnson. MK and RC are employees of Janssen. SKK received research funding from AbbVie, Amgen, Allogene, AstraZeneca, Bristol Myers Squibb, CARsgen, GlaxoSmithKline, Janssen, Novartis, Roche-Genentech, Takeda, Regeneron, and Molecular Templates; participated in consulting or an advisory board (with no personal payments) for AbbVie, Amgen, Bristol Myers Squibb, Janssen, Roche-Genentech, Takeda, AstraZeneca, bluebird bio, Epizyme, Secura Bio, Monte Rosa Therapeutics, Trillium, Loxo Oncology, K36, Sanofi, and Arcellx; and participated in consulting or an advisory board (with personal payments) for Oncopeptides, BeiGene, Antengene, and GLH Pharma., (© 2025. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2025
- Full Text
- View/download PDF
8. Benefit of the Omental Flap in Breast Reconstruction for Oncoplastic Treatment: A Systematic Review and Single-Arm Meta-Analysis.
- Author
-
Yang L, Tao C, Yan Y, Pan L, Li C, Jin X, Kong J, and Wu Z
- Abstract
Background: Oncoplastic surgery for breast cancer patients poses the challenge of achieving optimal aesthetic outcomes without increasing the risk of complications. Omental flap has emerged as n reconstructive option in breast surgery, yet the efficacy and safety of large omental flaps remain uncertain. This study aims to conduct a systematic review and single-arm meta-analysis to comprehensively evaluate the effectiveness, safety, and cosmetic outcomes of large omental flap breast reconstruction, providing updated evidence for clinical practice., Methods: We systematically searched Embase, PubMed, Cochrane Library, and CNKI databases until August 2023. Studies were screened using inclusion and exclusion criteria. The quality of each study was evaluated with the Newcastle-Ottawa Scale. Meta-analysis was performed using R version 4.2.0., Results: A total of 22 studies with 1031 patients were reviewed and analyzed. Meta-analysis results showed that hematoma and seroma rate were 4.1% (95%CI 0.8-8.8, P < 0.01). The pooled cosmetic outcomes demonstrated excellent rate 56.7% (95%CI 46.4-69.3, P < 0.01), good rate 7.9% (95%CI 3.9-11.9, P < 0.01), fair rate 28.7% (95%CI 19.9-37.5, P < 0.01). The pooled blood loss was 110.74 ml (95%CI: 72.33-149.14, P = 0), hospital stays was 7.27 (95%CI 5.65-8.89, P < 0.01), the time of omental flap harvest was 65.63 min (95%CI 59.95-71.32, P < 0.01), the time for surgery was 240.87 min (95%CI 5.65- 8.89, P = 0)., Conclusions: This meta-analysis indicates that the omental flap in breast reconstruction is safe and effective with good cosmetic outcomes and a low incidence of complications. The study highlights the benefits of comprehensive assessment, warranting further investigation through high-quality studies and long-term follow-up., Level of Evidence Iii: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 ., Competing Interests: Declarations. Conflict of interest: The authors declare that they have no conflict of interest. Statement of Human and Animal Rights: This article does not contain any studies with human participants or animals performed by any of the authors Informed Consent: For this type of study, informed consent is not required., (© 2024. Springer Science+Business Media, LLC, part of Springer Nature and International Society of Aesthetic Plastic Surgery.)
- Published
- 2024
- Full Text
- View/download PDF
9. Associations Among Adolescents' Mindfulness, Sympathy, Cognitive Empathy, and Sibling Relationships.
- Author
-
Barata, Özge, Acar, Ibrahim H., and Bostancı, Selen
- Subjects
SIBLINGS ,EMPATHY ,MINDFULNESS ,SYMPATHY ,TEENAGERS - Abstract
In the current study, we examined the direct and indirect paths from mindfulness to adolescents' sibling relationships through their cognitive empathy and sympathy. The sample consisted of 220 adolescents (50.9 % female) between age of 13 and 17 years (M = 15.86, SD = 0.91). Participants reported their mindfulness (acceptance and awareness), cognitive empathy and sympathy, and sibling relationships. The parallel mediation model revealed that mindful awareness and acceptance predicted kindness, involvement, and empathy within sibling relationships through sympathy. In addition, there was a significant indirect effect of mindful awareness to empathy in sibling relationships through cognitive empathy. Findings provided information regarding the importance of indirect contributions of mindfulness to sibling relationships through cognitive empathy and sympathy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. The Oncogenic Lipid Sphingosine-1-Phosphate Impedes the Phagocytosis of Tumor Cells by M1 Macrophages in Diffuse Large B Cell Lymphoma.
- Author
-
Perry, Tracey A., Masand, Navta, Vrzalikova, Katerina, Pugh, Matthew, Wei, Wenbin, Hollows, Robert, Bouchalova, Katerina, Nohtani, Mahdi, Fennell, Eanna, Bouchal, Jan, Kearns, Pamela, and Murray, Paul G.
- Subjects
RITUXIMAB ,PHAGOCYTOSIS ,MACROPHAGES ,B cell lymphoma ,MONOCLONAL antibodies ,IMMUNONUTRITION diet ,RESEARCH funding ,CELL lines ,SPHINGOSINE-1-phosphate ,CHEMICAL inhibitors - Abstract
Simple Summary: Diffuse large B cell lymphoma (DLBCL), the most common form of non-Hodgkin lymphoma, is clinically aggressive and associated with poor patient outcomes. Antagonists of the small oncogenic lipid sphingosine-1-phosphate (S1P) are already in the clinic and have been suggested to have therapeutic potential in DLBCL. We have studied the impact of S1P signaling on the recruitment of macrophages and their phagocytic functions following the treatment of DLBCL cells with CD20-targeting antibodies. We have shown that tumor-derived S1P is a major chemoattractant for monocytes and macrophages, both in vitro and in animal models of DLBCL, an effect mediated by the S1P receptor S1PR1. However, S1P also robustly inhibited the phagocytosis of antibody-treated tumor cells by M1 macrophages. Future experiments could be directed toward investigating the therapeutic effects of blocking S1P–S1PR1 signaling in combination with chemotherapy and CD20-targeting antibodies. Background: A total of 30–40% of diffuse large B cell lymphoma (DLBCL) patients will either not respond to the standard therapy or their disease will recur. The first-line treatment for DLBCL is rituximab and combination chemotherapy. This treatment involves the chemotherapy-induced recruitment of tumor-associated macrophages that recognize and kill rituximab-opsonized DLBCL cells. However, we lack insights into the factors responsible for the recruitment and functionality of macrophages in DLBCL tumors. Methods: We have studied the effects of the immunomodulatory lipid sphingosine-1-phosphate (S1P) on macrophage activity in DLBCL, both in vitro and in animal models. Results: We show that tumor-derived S1P mediates the chemoattraction of both monocytes and macrophages in vitro and in animal models, an effect that is dependent upon the S1P receptor S1PR1. However, S1P inhibited M1 macrophage-mediated phagocytosis of DLBCL tumor cells opsonized with the CD20 monoclonal antibodies rituximab and ofatumumab, an effect that could be reversed by an S1PR1 inhibitor. Conclusions: Our data show that S1P signaling can modulate macrophage recruitment and tumor cell killing by anti-CD20 monoclonal antibodies in DLBCL. The administration of S1PR1 inhibitors could enhance the phagocytosis of tumor cells and improve outcomes for patients. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Amino acid profiles: exploring their diagnostic and pathophysiological significance in hypertension.
- Author
-
Alqudah, Abdelrahim, Qnais, Esam, Wedyan, Mohammed, Awali, Ayat, Bseiso, Yousra, and Gammoh, Omar
- Abstract
Hypertension, a major contributor to cardiovascular morbidity, is closely linked to amino acid metabolism. Amino acids, particularly branched-chain amino acids (BCAAs) and aromatic amino acids (AAAs), may play pivotal roles in the pathogenesis and potential management of hypertension. This review investigated the relationships between amino acid profiles, specifically BCAAs and AAAs, and hypertension, and examined their potential as diagnostic and therapeutic targets. An in-depth analysis was conducted on studies highlighting the associations of specific amino acids such as arginine, glycine, proline, glutamine, and the BCAAs and AAAs with hypertension. BCAAs and AAAs, alongside other amino acids like arginine, glycine, and proline, showed significant correlations with hypertension. These amino acids influence multiple pathways including nitric oxide synthesis, vascular remodeling, and neurotransmitter production, among others. Distinct amino acid profiles were discerned between hypertensive and non-hypertensive individuals. Amino acid profiling, particularly the levels of BCAAs and AAAs, offers promising avenues in the diagnostic and therapeutic strategies for hypertension. Future studies are crucial to confirm these findings and to delineate amino acid-based interventions for hypertension treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Intravital imaging of the functions of immune cells in the tumor microenvironment during immunotherapy.
- Author
-
Xuwen Peng, Yuke Wang, Jie Zhang, Zhihong Zhang, and Shuhong Qi
- Subjects
TUMOR microenvironment ,IMMUNOTHERAPY ,CELL physiology ,IMMUNE response ,OPTICAL images - Abstract
Cancer immunotherapy has developed rapidly in recent years and stands as one of the most promising techniques for combating cancer. To develop and optimize cancer immunotherapy, it is crucial to comprehend the interactions between immune cells and tumor cells in the tumor microenvironment (TME). The TME is complex, with the distribution and function of immune cells undergoing dynamic changes. There are several research techniques to study the TME, and intravital imaging emerges as a powerful tool for capturing the spatiotemporal dynamics, especially the movement behavior and the immune function of various immune cells in real physiological state. Intravital imaging has several advantages, such as high spatio-temporal resolution, multicolor, dynamic and 4D detection, making it an invaluable tool for visualizing the dynamic processes in the TME. This review summarizes the workflow for intravital imaging technology, multi-color labeling methods, optical imaging windows, methods of imaging data analysis and the latest research in visualizing the spatiotemporal dynamics and function of immune cells in the TME. It is essential to investigate the role played by immune cells in the tumor immune response through intravital imaging. The review deepens our understanding of the unique contribution of intravital imaging to improve the efficiency of cancer immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
13. Breakthroughs in Cancer Immunotherapy: An Overview of T Cell, NK Cell, Mφ, and DC-Based Treatments.
- Author
-
Lee, Sunyoung and Kim, Tae-Don
- Subjects
KILLER cells ,CHIMERIC antigen receptors ,CYTOKINE release syndrome ,CANCER treatment ,GRAFT versus host disease ,T cells ,CYTOTOXIC T cells - Abstract
Efforts to treat cancer using chimeric antigen receptor (CAR)-T therapy have made astonishing progress and clinical trials against hematopoietic malignancies have demonstrated their use. However, there are still disadvantages which need to be addressed: high costs, and side effects such as Graft-versus-Host Disease (GvHD) and Cytokine Release Syndrome (CRS). Therefore, recent efforts have been made to harness the properties of certain immune cells to treat cancer—not just T cells, but also natural killer (NK) cells, macrophages (Mφ), dendritic cells (DC), etc. In this paper, we will introduce immune cell-based cellular therapies that use various immune cells and describe their characteristics and their clinical situation. The development of immune cell-based cancer therapy fully utilizing the unique advantages of each and every immune cell is expected to enhance the survival of tumor patients owing to their high efficiency and fewer side effects. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
14. Impact of daratumumab on stem cell mobilization and collection, engraftment and early post-transplant complications among multiple myeloma patients undergoing autologous stem cell transplantation.
- Author
-
Eleutherakis Papaiakovou, Evangelos, Terpos, Evangelos, Kanellias, Nikolaos, Migkou, Magdalini, Gavriatopoulou, Maria, Ntanasis-Stathopoulos, Ioannis, Fotiou, Despoina, Malandrakis, Panagiotis, Theodorakakou, Foteini, Spiliopoulou, Vasiliki, Kostopoulos, Ioannis V., Tsitsiloni, Ourania, Tsirigotis, Panagiotis, Dimopoulos, Meletios-Athanasios, and Kastritis, Efstathios
- Subjects
STEM cell transplantation ,DARATUMUMAB ,STEM cells ,MULTIPLE myeloma - Abstract
Autologous stem cell transplantation (ASCT) remains a standard therapy for multiple myeloma (MM) patients. Our study aimed to assess the impact of daratumumab-containing induction on stem cell (SC) mobilization, apheresis and hospitalization. We evaluated 200 newly diagnosed MM patients that were mobilized for SC collection and which received induction with (N = 40) or without daratumumab (N = 160). Dara group patients required more frequent use of plerixafor, larger collection volumes, and had lower SC yield. 87.5% (35/40) of dara group patients achieved the planned yield of ≥ 5 × 10^6 CD34+/kg for at least one transplant compared to 96.2% (154/160) of patients in the non-dara group. Dara group patients had delayed hematopoietic recovery (11 vs 10 days for PMN > 0.5 × 10E9/l), required more transfusions (4 vs 2 plts), prolonged hospitalization (20 vs 18 days), more febrile episodes and prolonged antibiotic administration. Despite daratumumab effect patients finally achieved a successful stem cell collection and proceeded to transplant. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. Safety and Efficacy of Subcutaneous Daratumumab in Systemic AL Amyloidosis.
- Author
-
Hughes, Michael Sang and Lentzsch, Suzanne
- Subjects
IMMUNOGLOBULIN light chains ,PLASMA cells ,DARATUMUMAB ,AMYLOIDOSIS ,MONOCLONAL antibodies - Abstract
Introduction: Systemic AL amyloidosis, a plasma cell dyscrasia, is characterized by the production of misfolded immunoglobulin light chain. These misfolded proteins aggregate into amyloid fibrils and deposit throughout the body, resulting in widespread organ dysfunction and ultimately death. Achieving rapid and maximal elimination of the plasma cell clone is crucial to long-term survival. Daratumumab, an anti-CD38 monoclonal antibody delivered intravenously, has been swiftly incorporated into standard first-line treatment regimens. A novel formulation of daratumumab has been developed that can be injected subcutaneously.Areas Covered: As a retrospective qualitative review of prior publications involving daratumumab, this work briefly summarizes the existing data regarding the safety and efficacy of subcutaneous (SC) daratumumab, compared to intravenous (IV) daratumumab. SC daratumumab appears to deliver the same disease benefit as IV daratumumab to patients with decreased infusion-related reactions (IRRs), decreased time for administration, and similar rates of adverse events (AEs) intrinsically related to daratumumab.Expert Opinion: SC daratumumab is preferred over IV daratumumab, but the clinical situation ultimately should determine route of administration. Further investigation into cost-effectiveness benefit is warranted. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
16. Daratumumab, bortezomib, melphalan, and prednisone versus bortezomib, melphalan, and prednisone alone in transplant-ineligible Asian patients with newly diagnosed multiple myeloma: final analysis of the phase 3 OCTANS Study.
- Author
-
Fu W, Bang SM, Huang H, Kim K, Li W, An G, Lee JJ, Cai Z, Jin J, Wang Y, Chim CS, Carson R, Liu R, Zhao M, Chen X, Cui C, Hou J, and Wang J
- Abstract
The superiority and tolerability of daratumumab plus bortezomib/melphalan/prednisone (D-VMP) versus bortezomib/melphalan/prednisone (VMP) in transplant-ineligible patients with newly diagnosed multiple myeloma (NDMM) was previously described in the global phase 3 ALCYONE study. The primary analysis of the phase 3 OCTANS study further demonstrated the superiority and tolerability of D-VMP (n = 144) versus VMP (n = 71) in transplant-ineligible Asian patients with NDMM. The current analysis describes the final efficacy and safety outcomes for D-VMP versus VMP in OCTANS, with a follow-up of > 3 years. D-VMP demonstrated a benefit versus VMP with regard to the rate of very good partial response or better (80.1% vs. 47.3%), median progression-free survival (38.7 vs. 19.2 months), median time to next treatment (46.8 vs. 20.6 months), rate of complete response or better (46.6% vs. 18.9%), median duration of response (41.3 vs. 18.5 months), achievement of minimal residual disease (MRD) negativity (40.4% vs. 10.8%), and sustained MRD negativity for ≥ 12 months (24.7% vs. 1.4%) and ≥ 18 months (15.1% vs. 1.4%). Median progression-free survival was longer among patients who achieved MRD negativity and sustained MRD negativity. The progression-free survival benefit observed with D-VMP was preserved across most clinically relevant subgroups, including patients with high-risk cytogenetics. No new safety concerns were identified with extended follow-up. This final analysis of OCTANS continues to demonstrate a clinical benefit for D-VMP versus VMP in transplant-ineligible Asian patients with NDMM, consistent with the global ALCYONE study, and supports the use of daratumumab combinations in this population. Trial registration: ClinicalTrials.gov Identifier NCT03217812 submitted July 13, 2017., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
17. DSE inhibits melanoma progression by regulating tumor immune cell infiltration and VCAN.
- Author
-
Xia, Lin, Feng, Maoxiao, Ren, Yidan, Hao, Xiaodong, Jiao, Qinlian, Xu, QinChen, Wang, Yunshan, Wang, Qin, and Gong, Ningji
- Published
- 2023
- Full Text
- View/download PDF
18. Role of daratumumab in the frontline management of multiple myeloma: a narrative review.
- Author
-
Yadav, Sanjeev, Gundeti, Sadashivudu, Bhave, Abhay, Deb, Uttiya, Dixit, Jitendra, and Mishra, Kundan
- Published
- 2023
- Full Text
- View/download PDF
19. A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma.
- Author
-
Candelaria, Pierre V., Nava, Miguel, Daniels-Wells, Tracy R., and Penichet, Manuel L.
- Subjects
IN vitro studies ,IMMUNOGLOBULINS ,IN vivo studies ,ANIMAL experimentation ,ANTINEOPLASTIC agents ,MACROPHAGES ,GENE expression ,MOLECULAR biology ,RESEARCH funding ,MULTIPLE myeloma ,DRUG development ,CELL lines ,ANTIGENS ,MONOCYTES ,MICE ,PHARMACODYNAMICS - Abstract
Simple Summary: Multiple myeloma (MM) is blood cancer of plasma cells. Plasma cells are white blood cells, which are part of the immune system. Malignant plasma cells are found in the bone marrow and are difficult to treat. There have been many new therapies developed in recent years, but the disease is still considered incurable. Some of the treatments for MM are antibodies of the IgG class. We have developed a fully human antibody of another antibody class, IgE, targeting the CD38 molecule that is expressed on the surface of several cancers including MM. Our goal is to use the anti-CD38 IgE antibody to recruit different types of immune cells to kill malignant cells. In this article, we report that the antibody shows anti-cancer effects against MM cells. Thus, this IgE antibody should be further explored as a novel treatment for MM. Multiple myeloma (MM) is an incurable malignancy of plasma cells and the second most common hematologic malignancy in the United States. Although antibodies in clinical cancer therapy are generally of the IgG class, antibodies of the IgE class have attractive properties as cancer therapeutics, such as their high affinity for Fc receptors (FcεRs), the low serum levels of endogenous IgE allowing for less competition for FcR occupancy, and the lack of inhibitory FcRs. Importantly, the FcεRs are expressed on immune cells that elicit antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), and/or antigen presentation such as mast cells, eosinophils, macrophages, and dendritic cells. We now report the development of a fully human IgE targeting human CD38 as a potential MM therapy. We targeted CD38 given its high and uniform expression on MM cells. The novel anti-CD38 IgE, expressed in mammalian cells, is properly assembled and secreted, exhibits the correct molecular weight, binds antigen and the high affinity FcεRI, and induces degranulation of FcεRI expressing cells in vitro and also in vivo in transgenic BALB/c mice expressing human FcεRIα. Moreover, the anti-CD38 IgE induces ADCC and ADCP mediated by monocytes/macrophages against human MM cells (MM.1S). Importantly, the anti-CD38 IgE also prolongs survival in a preclinical disseminated xenograft mouse model using SCID-Beige mice and human MM.1S cells when administered with human peripheral blood mononuclear cells (PBMCs) as a source of monocyte effector cells. Our results suggest that anti-CD38 IgE may be effective in humans bearing MM and other malignancies expressing CD38. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. Switching to daratumumab SC from IV is safe and preferred by patients with multiple myeloma.
- Author
-
Mateos, Maria-Victoria, Rigaudeau, Sophie, Basu, Supratik, Spicka, Ivan, Schots, Rik, Wrobel, Tomasz, Cook, Gordon, Beksac, Meral, Gries, Katharine S, Kudva, Anupa, Tromp, Brenda, Van Rampelbergh, Rian, Pei, Huiling, Wroblewski, Susan, Carson, Robin, Delioukina, Maria, and White, Darrell
- Subjects
THERAPEUTIC use of monoclonal antibodies ,GENERIC drug substitution ,RANDOMIZED controlled trials ,RESEARCH funding ,DESCRIPTIVE statistics ,MULTIPLE myeloma ,STATISTICAL sampling ,PATIENT safety - Abstract
Introduction: Two phase 3 studies demonstrated superior efficacy of intravenous daratumumab (DARA IV) plus bortezomib/melphalan/prednisone (ALCYONE) or lenalidomide/dexamethasone (Rd; MAIA) versus standard-of-care regimens for transplant-ineligible newly diagnosed multiple myeloma. In these studies, patients could switch from DARA IV to subcutaneous daratumumab (DARA SC) while receiving daratumumab monotherapy in ALCYONE (as of Cycle 11) or daratumumab plus Rd in MAIA. The phase 3 COLUMBA study demonstrated noninferiority of DARA SC to DARA IV. DARA SC reduced administration time, allowing patients to spend less time in healthcare settings, a relevant practical consideration for patient care in the COVID-19 pandemic/settings of limited healthcare resources. Methods: DARA SC 1800 mg was administered every 4 weeks, per approved dosing schedules. We evaluated safety and patient-reported experience (ALCYONE only) among patients who switched from DARA IV to DARA SC. Results: Fifty-seven patients in ALCYONE and 135 in MAIA switched to DARA SC. Three (2.2%; MAIA) patients reported injection-site reactions, all of which were mild. No infusion-related reactions occurred with DARA SC. In ALCYONE, >80% of patients preferred DARA SC over DARA IV. Grade 3/4 treatment-emergent adverse events (TEAEs) occurred in 5.3% of patients in ALCYONE and 25.9% in MAIA; one (0.7%; MAIA) patient experienced a TEAE with an outcome of death. Conclusion: For transplant-ineligible newly diagnosed multiple myeloma, DARA SC (monotherapy/with Rd) was safe and preferred over DARA IV. ClinicalTrials.gov, NCT02195479/NCT02252172. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
21. Clinical efficacy of retreatment of daratumumab‐based therapy (D2) in daratumumab‐refractory multiple myeloma.
- Author
-
Abdallah, Al‐Ola, Mahmoudjafari, Zahra, Ahmed, Nausheen, Cui, Wei, Shune, Leyla, McGuirk, Joseph, Mohan, Meera, Mohyuddin, Ghulam Rehman, Afrough, Aimaz, Alkharabsheh, Omar, and Atrash, Shebli
- Subjects
MULTIPLE myeloma ,DARATUMUMAB ,PROTEASOME inhibitors ,EXTRAMEDULLARY diseases ,DATABASES ,PLASMACYTOMA - Abstract
Daratumumab demonstrates activity as a single agent and in combination with either immunomodulatory agents (IMiDs) or proteasome inhibitors (PIs) in relapsed refractory multiple myeloma (RRMM). However, little is known about the benefit of daratumumab retreatment in daratumumab‐refractory MM. This study aimed to analyze the clinical efficacy of daratumumab‐based retreatment (D2) in patients who are daratumumab refractory MM. Retrospectively, we identified 43 RRMM patients from a single‐center database review. The median age was 65 years, 42% patients had high‐risk cytogenetics, and 23% had an extramedullary disease, while the median time between D2 and prior daratumumab was 1 (0.25–39) month. All D2 patients received combination therapy with either pomalidomide, carfilzomib, bortezomib, or lenalidomide. The response rate, median progression‐free, and overall survival were 49%, 7.97 and 32.6 months, respectively. Our study raises the possibility of re‐utilizing daratumumab in combination with different classes of anti‐myeloma drugs to generate responses in RRMM patients who are daratumumab‐refractory. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
22. Clinical Pharmacokinetics and Pharmacodynamics of Daratumumab.
- Author
-
Kim, Kyeongmin and Phelps, Mitch A.
- Subjects
ALBUMINS ,DARATUMUMAB ,IMMUNOGLOBULIN G ,CANCER cells ,PHARMACOKINETICS ,PHARMACODYNAMICS - Abstract
Daratumumab is a fully human, monoclonal immunoglobulin G1 and a first-in-class CD38-targeting drug approved by the US Food and Drug Administration for the treatment of patients with relapsed/refractory and newly diagnosed multiple myeloma or newly diagnosed light-chain amyloidosis. CD38 is heavily expressed on malignant myeloma cells, and daratumumab exerts anti-myeloma activity via immune-mediated mechanisms, direct induction of apoptosis, and immunomodulation. Daratumumab is used as monotherapy or in combination with standard-of-care myeloma therapies, including proteasome inhibitors, immunomodulatory agents, DNA-alkylating agents, and corticosteroids. Following an intravenous infusion, daratumumab exhibits nonlinear pharmacokinetics (PK), as clearance decreases with higher doses and over time because of target-mediated effects. Dosing schedules vary depending on indications and co-administered drugs, but generally daratumumab is administered weekly for 6–9 weeks followed by a less frequent dosing regimen, once every 2–4 weeks. Daratumumab exposure is strongly correlated with efficacy, and the exposure–efficacy relationship follows a maximal effect model, whereas exposure is not correlated with safety endpoints. The approved dose of 16 mg/kg of daratumumab results in the saturation of 99% of the target at the end of weekly dosing in most patients, and high target saturation is maintained over time during the less frequent dosing schedule. Infusion-related reactions are frequently observed in patients given daratumumab, particularly with the first infusion, thus prompting long durations of infusion (~ 7 h) and splitting of the first dose across 2 days. This led to the development of a subcutaneous delivery formulation for daratumumab (Dara-SC). Dara-SC provides a similar efficacy and safety profile to intravenous daratumumab (Dara-IV) but has a much lower rate of infusion-related reactions and a shorter infusion time. Exposure–response relationships for efficacy and safety endpoints were similar between Dara-SC and Dara-IV, and co-administered drugs with either Dara-IV or Dara-SC do not significantly affect daratumumab PK. Except for baseline myeloma type and albumin level, none of the other investigated disease and patient characteristics (renal/hepatic function, age, sex, race, weight, Eastern Cooperative Oncology Group performance status) was identified to have clinically relevant effects on exposure to daratumumab monotherapy or combination therapy regimens. Dara-IV exposure was significantly lower in patients with immunoglobulin G myeloma compared with patients with non-immunoglobulin G myeloma (p < 0.0001) and in patients with a lower albumin level, whereas the overall response rate was similar regardless of the myeloma type and albumin level. Daratumumab dose adjustment is not currently recommended based on disease and patient characteristics. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
23. Bortezomib, Melphalan, and Prednisone With or Without Daratumumab in Transplant-ineligible Asian Patients With Newly Diagnosed Multiple Myeloma: The Phase 3 OCTANS Study.
- Author
-
Fu, Weijun, Bang, Soo-Mee, Huang, Honghui, Kim, Kihyun, Li, Wei, An, Gang, Lee, Je-Jung, Cai, Zhen, Jin, Jie, Wang, Yafei, Lin, Tung-Liang, Chim, Chor Sang, Qi, Ming, Wang, Jianping, Lu, Xiaolin, Song, Yang, Jia, Bin, Yang, Xue, Liu, Wenyu, and Zhou, Tianyuan
- Published
- 2023
- Full Text
- View/download PDF
24. The battle within: cell death by phagocytosis in cancer.
- Author
-
Zhou L, Fan S, Zhang W, Gong Z, Wang D, and Tang D
- Abstract
The process by which living cells are phagocytosed and digested to death is called cell death by phagocytosis, a term that has just recently been generalized and redefined. It is characterized by the phagocytosis of living cells and the cessation of cell death by phagocytosis. Phagocytosis of dead cells is a widely discussed issue in cancer, cell death by phagocytosis can stimulate phagocytosis and stimulate adaptive immunity in tumors, and at the same time, do not-eat-me signaling is an important site for cancer cells to evade recognition by phagocytes. Therefore, we discuss in this review cell death by phagocytosis occurring in cancer tissues and emphasize the difference between this new concept and the phagocytosis of dead tumor cells. Immediately thereafter, we describe the mechanisms by which cell death by phagocytosis occurs and how tumors escape phagocytosis. Finally, we summarize the potential clinical uses of cell death by phagocytosis in tumor therapy and strive to provide ideas for tumor therapy., (© 2024. The Author(s), under exclusive licence to Federación de Sociedades Españolas de Oncología (FESEO).)
- Published
- 2024
- Full Text
- View/download PDF
25. Enhancing the therapeutic activity of hyperimmune IgG against chikungunya virus using FcγRIIIa affinity chromatography.
- Author
-
Fox, Julie M., Roy, Vicky, Gunn, Bronwyn M., Bolton, Glen R., Fremont, Daved H., Alter, Galit, Diamond, Michael S., and Boesch, Austin W.
- Subjects
CHIKUNGUNYA virus ,AFFINITY chromatography ,IMMUNOGLOBULIN G ,IMMUNE response ,BIOLOGICAL assay - Abstract
Introduction: Chikungunya virus (CHIKV) is a re-emerging mosquito transmitted alphavirus of global concern. Neutralizing antibodies and antibody Fc-effector functions have been shown to reduce CHIKV disease and infection in animals. However, the ability to improve the therapeutic activity of CHIKV-specific polyclonal IgG by enhancing Fc-effector functions through modulation of IgG subclass and glycoforms remains unknown. Here, we evaluated the protective efficacy of CHIKV-immune IgG enriched for binding to Fc-gamma receptor IIIa (FcgRIIIa) to select for IgG with enhanced Fc effector functions. Methods: Total IgG was isolated from CHIKV-immune convalescent donors with and without additional purification by FcgRIIIa affinity chromatography. The enriched IgG was characterized in biophysical and biological assays and assessed for therapeutic efficacy during CHIKV infection in mice. Results: FcgRIIIa-column purification enriched for afucosylated IgG glycoforms. In vitro characterization showed the enriched CHIKV-immune IgG had enhanced human FcgRIIIa and mouse FcgRIV affinity and FcgR-mediated effector function without reducing virus neutralization in cellular assays. When administered as postexposure therapy in mice, CHIKV-immune IgG enriched in afucosylated glycoforms promoted reduction in viral load. Discussion: Our study provides evidence that, in mice, increasing Fc engagement of FcgRs on effector cells, by leveraging FcgRIIIa-affinity chromatography, enhanced the antiviral activity of CHIKV-immune IgG and reveals a path to produce more effective therapeutics against these and potentially other emerging viruses. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
26. Anti-Tumor Strategies by Harnessing the Phagocytosis of Macrophages.
- Author
-
Li, Si-Yuan, Guo, Yong-Lin, Tian, Jia-Wen, Zhang, He-Jing, Li, Rui-Fang, Gong, Ping, and Yu, Zi-Li
- Subjects
PHAGOCYTOSIS ,MACROPHAGES ,CELLULAR signal transduction ,NANOMEDICINE - Abstract
Simple Summary: Macrophages are the "big eaters" of the immune system who are in charge of engulfing undesirable substances. Macrophages are vital for the human body as they are instrumental in developing organisms, regulating immune responses, and maintaining a relatively stable internal environment. When the phagocytic capacity of macrophages is impaired, the body is prone to disease. In the context of tumors, tumor cells have their ways to escape from macrophage-mediated phagocytosis. They masquerade as healthy cells by expressing "don't eat me" signals to fool macrophages and turn the initially anti-tumoral macrophages against the human body, which results in reduced macrophage-mediated phagocytosis. Hence, promoting the phagocytosis of macrophages is an important approach to improving the efficacy of anti-tumor treatment. In this review, we introduced the underlying mechanisms of macrophage-mediated phagocytosis and reviewed the recent progress in the area of application strategies on the basis of the phagocytosis mechanism. Macrophages are essential for the human body in both physiological and pathological conditions, engulfing undesirable substances and participating in several processes, such as organism growth, immune regulation, and maintenance of homeostasis. Macrophages play an important role in anti-bacterial and anti-tumoral responses. Aberrance in the phagocytosis of macrophages may lead to the development of several diseases, including tumors. Tumor cells can evade the phagocytosis of macrophages, and "educate" macrophages to become pro-tumoral, resulting in the reduced phagocytosis of macrophages. Hence, harnessing the phagocytosis of macrophages is an important approach to bolster the efficacy of anti-tumor treatment. In this review, we elucidated the underlying phagocytosis mechanisms, such as the equilibrium among phagocytic signals, receptors and their respective signaling pathways, macrophage activation, as well as mitochondrial fission. We also reviewed the recent progress in the area of application strategies on the basis of the phagocytosis mechanism, including strategies targeting the phagocytic signals, antibody-dependent cellular phagocytosis (ADCP), and macrophage activators. We also covered recent studies of Chimeric Antigen Receptor Macrophage (CAR-M)-based anti-tumor therapy. Furthermore, we summarized the shortcomings and future applications of each strategy and look into their prospects with the hope of providing future research directions for developing the application of macrophage phagocytosis-promoting therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
27. Population pharmacokinetics and exposure–response analyses of daratumumab plus pomalidomide/dexamethasone in relapsed or refractory multiple myeloma.
- Author
-
Dosne, Anne‐Gaelle, Li, Xia, Luo, Man Melody, Nnane, Ivo, Dimopoulos, Meletios A., Terpos, Evangelos, Sonneveld, Pieter, Kampfenkel, Tobias, Carson, Robin, Amin, Himal, Perez Ruixo, Juan, Zhou, Honghui, Sun, Yu‐Nien, and Xu, Yan
- Subjects
DARATUMUMAB ,PHARMACOKINETICS ,MULTIPLE myeloma ,OVERALL survival ,DEXAMETHASONE ,INTRAVENOUS therapy - Abstract
Aim: A population pharmacokinetic (PPK) model was developed to characterize pharmacokinetics (PK) of subcutaneous or intravenous daratumumab administration in a new indication (i.e., combination with pomalidomide and dexamethasone [D‐Pd] in patients with relapsed or refractory multiple myeloma [RRMM]). Analyses were conducted to explore exposure–response (E‐R) relationships for efficacy and select treatment‐emergent adverse events (TEAEs). Methods: The PPK analysis included pooled data from the D‐Pd cohorts of the phase 3 APOLLO and phase 1b EQUULEUS studies. Covariates were evaluated in the PPK model. Model‐predicted exposures to daratumumab were compared between covariate subgroups of interest and used to investigate relationships between daratumumab exposure and efficacy and safety in APOLLO. Results: The PPK analysis included 1146 daratumumab PK samples from 239 patients (APOLLO, n = 140; EQUULEUS, n = 99). Observed concentration–time data of daratumumab were well described by a two‐compartment PPK model with first‐order absorption and parallel linear and nonlinear elimination pathways. Treatment with D‐Pd provided similar daratumumab PK characteristics versus historical daratumumab monotherapy. The E‐R dataset contained data from 290 APOLLO patients (D‐Pd, n = 140; Pd, n = 150). The PK–efficacy relationship of daratumumab supported improved progression‐free survival for patients in the D‐Pd group vs. the Pd group. Additionally, TEAEs did not increase with increasing PK exposure in the D‐Pd group. Conclusions: The PPK and E‐R analyses support the daratumumab subcutaneous 1800 mg dosing regimen in combination with Pd for treatment of patients with RRMM. No dose adjustment is recommended in this indication for any of the investigated factors, none of which had clinically relevant effects on daratumumab PK. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
28. Extracellular Vesicles (EVs) in Tumor Diagnosis and Therapy.
- Author
-
Tan, Mingdian, Ge, Yizhi, Wang, Xiaogang, Wang, Yan, Liu, Yi, He, Feng, and Teng, Hongqi
- Subjects
EXTRACELLULAR vesicles ,TUMOR diagnosis ,MEDICAL research ,CLINICAL medicine ,TUMOR microenvironment ,NANOMEDICINE ,PROGRAMMED cell death 1 receptors - Abstract
In recent years, extracellular vesicles (EVs) have gained significant attention due to their tremendous potential for clinical applications. EVs play a crucial role in various aspects, including tumorigenesis, drug resistance, immune escape, and reconstruction of the tumor microenvironment. Despite the growing interest in EVs, many questions still need to be addressed before they can be practically applied in clinical settings. This paper aims to review EVs' isolation methods, structure research, the roles of EVs in tumorigenesis and their mechanisms in multiple types of tumors, their potential application in drug delivery, and the expectations for their future in clinical research. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
29. Daratumumab plus bortezomib, cyclophosphamide, and dexamethasone in Asian patients with newly diagnosed AL amyloidosis: subgroup analysis of ANDROMEDA.
- Author
-
Suzuki, Kenshi, Wechalekar, Ashutosh D., Kim, Kihyun, Shimazaki, Chihiro, Kim, Jin Seok, Ikezoe, Takayuki, Min, Chang-Ki, Zhou, Fude, Cai, Zhen, Chen, Xiaonong, Iida, Shinsuke, Katoh, Nagaaki, Fujisaki, Tomoaki, Shin, Ho-Jin, Tran, NamPhuong, Qin, Xiang, Vasey, Sandra Y., Tromp, Brenda, Weiss, Brendan M., and Comenzo, Raymond L.
- Subjects
ASIANS ,DARATUMUMAB ,IMMUNOGLOBULIN light chains ,AMYLOIDOSIS ,BORTEZOMIB - Abstract
Subcutaneous daratumumab plus bortezomib/cyclophosphamide/dexamethasone (VCd; D-VCd) improved outcomes versus VCd for patients with newly diagnosed immunoglobulin light-chain (AL) amyloidosis in the phase 3 ANDROMEDA study. We report a subgroup analysis of Asian patients (Japan; Korea; China) from ANDROMEDA. Among 388 randomized patients, 60 were Asian (D-VCd, n = 29; VCd, n = 31). At a median follow-up of 11.4 months, the overall hematologic complete response rate was higher for D-VCd versus VCd (58.6% vs. 9.7%; odds ratio, 13.2; 95% confidence interval [CI], 3.3–53.7; P < 0.0001). Six-month cardiac and renal response rates were higher with D-VCd versus VCd (cardiac, 46.7% vs. 4.8%; P = 0.0036; renal, 57.1% vs. 37.5%; P = 0.4684). Major organ deterioration progression-free survival (MOD-PFS) and major organ deterioration event-free survival (MOD-EFS) were improved with D-VCd versus VCd (MOD-PFS: hazard ratio [HR], 0.21; 95% CI, 0.06–0.75; P = 0.0079; MOD-EFS: HR, 0.16; 95% CI, 0.05–0.54; P = 0.0007). Twelve deaths occurred (D-VCd, n = 3; VCd, n = 9). Twenty-two patients had baseline serologies indicating prior hepatitis B virus (HBV) exposure; no patient experienced HBV reactivation. Although grade 3/4 cytopenia rates were higher than in the global safety population, the safety profile of D-VCd in Asian patients was generally consistent with the global study population, regardless of body weight. These results support D-VCd use in Asian patients with newly diagnosed AL amyloidosis. ClinicalTrials.gov Identifier: NCT03201965. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
30. Propargyl Amines: Versatile Building Blocks in Post‐Ugi Transformations.
- Author
-
Bhoraniya, Rinkal B. and Modha, Sachin G.
- Subjects
AMINES ,METAMORPHOSIS - Abstract
The Ugi reaction, a multicomponent reaction, allows diversity‐oriented synthesis Its importance is recognized by an exponential increase in the publications utilizing the post‐Ugi transformations as a strategy to build complex molecules via simple and sustainable processes in the recent literature. A second concept, alkyne activation through metal‐, acid‐, iodine‐catalysis and base‐mediated transformations, also leads to wonderful molecules in short and efficient synthetic routes. Combination of these two approaches via application of an alkyne‐containing component in the Ugi reaction brings the benefits of both protocols into one synthetic sequence. The propargyl amines come as an obvious choice in this context as they work wonderfully as an amine component in the Ugi reaction, while post‐Ugi alkyne activation has the potential to generate biologically interesting carbo‐ and hetero‐cyclic systems. Thus, one can compare the Ugi adduct with a pupa which has inherent property of metamorphosis into biologically interesting molecules. In this review, application of propargyl amines in the Ugi reaction is discussed with a focus on post‐Ugi transformations. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
31. Microbiota dysbiosis influences immune system and muscle pathophysiology of dystrophin‐deficient mice.
- Author
-
Farini, Andrea, Tripodi, Luana, Villa, Chiara, Strati, Francesco, Facoetti, Amanda, Baselli, Guido, Troisi, Jacopo, Landolfi, Annamaria, Lonati, Caterina, Molinaro, Davide, Wintzinger, Michelle, Gatti, Stefano, Cassani, Barbara, Caprioli, Flavio, Facciotti, Federica, Quattrocelli, Mattia, and Torrente, Yvan
- Abstract
Duchenne muscular dystrophy (DMD) is a progressive severe muscle‐wasting disease caused by mutations in DMD, encoding dystrophin, that leads to loss of muscle function with cardiac/respiratory failure and premature death. Since dystrophic muscles are sensed by infiltrating inflammatory cells and gut microbial communities can cause immune dysregulation and metabolic syndrome, we sought to investigate whether intestinal bacteria support the muscle immune response in mdx dystrophic murine model. We highlighted a strong correlation between DMD disease features and the relative abundance of Prevotella. Furthermore, the absence of gut microbes through the generation of mdx germ‐free animal model, as well as modulation of the microbial community structure by antibiotic treatment, influenced muscle immunity and fibrosis. Intestinal colonization of mdx mice with eubiotic microbiota was sufficient to reduce inflammation and improve muscle pathology and function. This work identifies a potential role for the gut microbiota in the pathogenesis of DMD. Synopsis: The susceptibility of DMD patients to inflammatory events cannot solely be explained by skeletal muscle genetic defects but rather favors a new paradigm linking development of chronic inflammation with a strict regulation between epigenetics factors and degenerative environment. Gut microbiota–specific alterations (dysbiosis) correlate with the dystrophic pathology in mdx mice, influencing muscle immunity and fibrosis.Dysbiotic mdx microbiota induces a decreased innate immune response and altered muscle metabolism.The study of the dysregulated immune system‐microbiota axis in mdx mice highlights the importance of microbiota as a potential target for therapeutic interventions. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
32. The molecular biology of peritoneal metastatic disease.
- Author
-
Bootsma, Sanne, Bijlsma, Maarten F, and Vermeulen, Louis
- Abstract
Peritoneal metastases are a common form of tumor cell dissemination in gastrointestinal malignancies. Peritoneal metastatic disease (PMD) is associated with severe morbidity and resistance to currently employed therapies. Given the distinct route of dissemination compared with distant organ metastases, and the unique microenvironment of the peritoneal cavity, specific tumor cell characteristics are needed for the development of PMD. In this review, we provide an overview of the known histopathological, genomic, and transcriptomic features of PMD. We find that cancers representing the mesenchymal subtype are strongly associated with PMD in various malignancies. Furthermore, we discuss the peritoneal niche in which the metastatic cancer cells reside, including the critical role of the peritoneal immune system. Altogether, we show that PMD should be regarded as a distinct disease entity, that requires tailored treatment strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
33. Relapsed/Refractory Multiple Myeloma: A Review of Available Therapies and Clinical Scenarios Encountered in Myeloma Relapse.
- Author
-
Bhatt, Parva, Kloock, Colin, and Comenzo, Raymond
- Subjects
MULTIPLE myeloma ,CANCER relapse ,STEM cell transplantation ,HUMAN cytogenetics ,PROTEASOME inhibitors ,EXTRAMEDULLARY diseases - Abstract
Multiple myeloma remains an incurable disease with the usual disease course requiring induction therapy, autologous stem cell transplantation for eligible patients, and long-term maintenance. Risk stratification tools and cytogenetic alterations help inform individualized therapeutic choices for patients in hopes of achieving long-term remissions with preserved quality of life. Unfortunately, relapses occur at different stages of the course of the disease owing to the biological heterogeneity of the disease. Addressing relapse can be complex and challenging as there are both therapy- and patient-related factors to consider. In this broad scoping review of available therapies in relapsed/refractory multiple myeloma (RRMM), we cover the pharmacologic mechanisms underlying active therapies such as immunomodulatory agents (IMiDs), proteasome inhibitors (PIs), monoclonal antibodies (mAbs), traditional chemotherapy, and Venetoclax. We then review the clinical data supporting the use of these therapies, organized based on drug resistance/refractoriness, and the role of autologous stem cell transplant (ASCT). Approaches to special situations during relapse such as renal impairment and extramedullary disease are also covered. Lastly, we look towards the future by briefly reviewing the clinical data supporting the use of chimeric antigen receptor (CAR-T) therapy, bispecific T cell engagers (BITE), and Cereblon E3 Ligase Modulators (CELMoDs). [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
34. Targeting Notch-Driven Cytokine Secretion: Novel Therapies for Triple Negative Breast Cancer.
- Author
-
Marini, Wanda, Wilson, Brooke E., and Reedijk, Michael
- Subjects
TRIPLE-negative breast cancer ,IMMUNE checkpoint inhibitors ,BREAST cancer ,NOTCH genes ,CYTOKINES - Abstract
Compared with other breast cancer subtypes, triple negative breast cancer (TNBC) is an aggressive malignancy with a high recurrence rate and reduced overall survival. Immune checkpoint inhibition (ICI) has shown modest results in this subgroup, highlighting the need for improved targeted therapeutic options. Notch is a defining feature of TNBC and drives the expression of interleukin-1 beta (IL1β) and C-C motif chemokine ligand 2 (CCL2). These cytokines are involved in the recruitment of tumor-associated macrophages (TAMs) to the tumor, resulting in immune evasion and tumor progression. Targeting Notch, IL1β or CCL2 may reduce TAM recruitment and resistance to ICI, illuminating the potential of combination immunotherapy in TNBC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
35. Aspirin increases chemosensitivity of colorectal cancer cells and inhibits the expression of toll-like receptor 4.
- Author
-
Ying, Jun, Zhou, Haiyang, Wang, Zhiguo, You, Qing, Chen, Junnan, Lu, Hao, and Zhang, Jian
- Subjects
COLORECTAL cancer ,TOLL-like receptors ,CANCER cells ,ASPIRIN ,COLON cancer prognosis ,COLON cancer - Abstract
Background: Chemotherapy resistance is an important bottleneck affecting the efficacy of chemotherapy in colon cancer. Therefore, improving the chemotherapy sensitivity of colorectal cancer cells is of great significance for improving the prognosis of patients with colon cancer. Methods: CCK-8 assay was employed to examine the cell viability of colorectal cancer cell lines. Realtime-PCR and western blot were used to explore toll-like receptor 4 (TLR4) expression in colorectal cancer cell lines. The functions of TLR4 in the stemness of the colorectal cancer cell lines were analyzed by infecting cells with lentivirus containing TLR4 siRNA. Results: We found that aspirin could effectively enhance the chemosensitivity of CT26 and HCT116 colorectal cancer cell lines. Aspirin can also inhibit the stemness of colorectal cancer cell including inhibiting the number of clone formation and reducing the volume and number of cell spheres and inducing the down-regulation of stemness-related genes. Besides that, aspirin also lead to down-regulation of TLR4 expression in colorectal cancer cells. The TLR4 positive colorectal cancer cells demonstrated a higher chemotherapy resistance potential than TLR4 negative colorectal cancer cells. In addition, the stemness of TLR4 positive colorectal cancer cells is stronger than TLR4 negative colorectal cancer cells. Conclusion: The results of our study indicate that aspirin increases chemosensitivity of colorectal cancer cells and inhibits the expression of toll-like receptor 4. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
36. Daratumumab, Bortezomib, and Dexamethasone versus Bortezomib and Dexamethasone in Chinese Patients With Relapsed or Refractory Multiple Myeloma: Updated Analysis of LEPUS.
- Author
-
Fu, Weijun, Li, Wei, Hu, Jianda, An, Gang, Wang, Yafei, Fu, Chengcheng, Chen, Lijuan, Jin, Jie, Cen, Xinan, Ge, Zheng, Cai, Zhen, Niu, Ting, Qi, Ming, Gai, Xue, Li, Qian, Liu, Weiping, Liu, Wenyu, Yang, Xue, Chen, Xi, and Lu, Jin
- Published
- 2023
- Full Text
- View/download PDF
37. Efficacy, Safety, and Challenges of CAR T-Cells in the Treatment of Solid Tumors.
- Author
-
Chen, Qiuqiang, Lu, Lingeng, and Ma, Wenxue
- Subjects
TUMOR treatment ,DRUG efficacy ,NEUROTOXICOLOGY ,SYNDROMES ,CELLULAR therapy ,TREATMENT effectiveness ,CYTOKINE release syndrome ,HEMATOLOGIC malignancies ,T cells ,IMMUNOTHERAPY ,PATIENT safety ,EVALUATION - Abstract
Simple Summary: Chimeric antigen receptor T cells (CAR T-cells) are engineered T cells that target tumor-associated antigens. CAR T-cell therapy is a novel developed immunotherapy initially for destroying hematological malignancies. Its great success in clinical practice of hematological malignancies encourages oncologists and scientists to use CAR T-cells for the treatment of solid cancers. However, the efficacy of CAR T-cells in solid tumors is not as good as expected in hematological malignancies. In this review, we summarized the efficacy, safety, and challenges of CAR T-cell therapy in the clinical management of solid tumors. We also discussed the potential strategies currently applied to improve the efficacy and safety of CAR T-cell therapy in solid tumors, and finally prospected the future study direction for CAR T-cell therapy. Immunotherapy has been the fifth pillar of cancer treatment in the past decade. Chimeric antigen receptor (CAR) T-cell therapy is a newly designed adoptive immunotherapy that is able to target and further eliminate cancer cells by engaging with MHC-independent tumor-antigens. CAR T-cell therapy has exhibited conspicuous clinical efficacy in hematological malignancies, but more than half of patients will relapse. Of note, the efficacy of CAR T-cell therapy has been even more disappointing in solid tumors. These challenges mainly include (1) the failures of CAR T-cells to treat highly heterogeneous solid tumors due to the difficulty in identifying unique tumor antigen targets, (2) the expression of target antigens in non-cancer cells, (3) the inability of CAR T-cells to effectively infiltrate solid tumors, (4) the short lifespan and lack of persistence of CAR T-cells, and (5) cytokine release syndrome and neurotoxicity. In combination with these characteristics, the ideal CAR T-cell therapy for solid tumors should maintain adequate T-cell response over a long term while sparing healthy tissues. This article reviewed the status, clinical application, efficacy, safety, and challenges of CAR T-cell therapies, as well as the latest progress of CAR T-cell therapies for solid tumors. In addition, the potential strategies to improve the efficacy of CAR T-cells and prevent side effects in solid tumors were also explored. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
38. Uptake of Plasmodium chabaudi hemozoin drives Kupffer cell death and fuels superinfections.
- Author
-
Hirako, Isabella C., Antunes, Maísa Mota, Rezende, Rafael Machado, Hojo-Souza, Natália Satchiko, Figueiredo, Maria Marta, Dias, Thomaz, Nakaya, Helder, Menezes, Gustavo Batista, and Gazzinelli, Ricardo Tostes
- Subjects
KUPFFER cells ,CELL death ,SUPERINFECTION ,PLASMODIUM ,FUEL cells - Abstract
Kupffer cells (KCs) are self-maintained tissue-resident macrophages that line liver sinusoids and play an important role on host defense. It has been demonstrated that upon infection or intense liver inflammation, KCs might be severely depleted and replaced by immature monocytic cells; however, the mechanisms of cell death and the alterations on liver immunity against infections deserves further investigation. We explored the impact of acute Plasmodium infection on KC biology and on the hepatic immune response against secondary infections. Similar to patients, infection with Plasmodium chabaudi induced acute liver damage as determined by serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) elevation. This was associated with accumulation of hemozoin, increased of proinflammatory response and impaired bacterial and viral clearance, which led to pathogen spread to other organs. In line with this, mice infected with Plasmodium had enhanced mortality during secondary infections, which was associated with increased production of mitochondrial superoxide, lipid peroxidation and increased free iron within KCs—hallmarks of cell death by ferroptosis. Therefore, we revealed that accumulation of iron with KCs, triggered by uptake of circulating hemozoin, is a novel mechanism of macrophage depletion and liver inflammation during malaria, providing novel insights on host susceptibility to secondary infections. Malaria can cause severe liver damage, along with depletion of liver macrophages, which can predispose individuals to secondary infections and enhance the chances of death. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
39. A REVIEW OF STRESS ON STUDENTS WITH ADHD. THE ROLE OF ICTS & MENDAL INTERVENTIONS TO IMPROVE PRODUCTIVITY.
- Author
-
Tsakiridou, Maria and Drigas, Athanasios
- Subjects
ATTENTION-deficit hyperactivity disorder ,NEURAL development ,CHRONIC diseases ,SCHOOL entrance age ,ANXIETY - Abstract
The Attention Deficit Hyperactivity Disorder (AD/HD) is the most common neurodevelopmental disorder of childhood and one of the more widespread chronic diseases that affect children school age, based on the DSM-IV. This study focuses on the influence of stress to the kids suffering from ADHD and the importance of other elements on it. This particular study was based on articles in scientific journals, with the purpose to investigate some of the factors that affect positively or negatively in the exacerbation of anxiety on children suffering from ADHD and how the prenatal and parental stress affects this condition. The results have shown that the environmental factors, nutrition, hormones, neurotransmitters and alternative techniques are directly related with the characteristics of the disorder and the ways of how to improve it. In the light of these findings, we came to the conclusion that we can intervene to the deficits of mainly socio-emotional functions through the above factors. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
40. Daratumumab plus lenalidomide, bortezomib and dexamethasone in newly diagnosed multiple myeloma: Analysis of vascular thrombotic events in the GRIFFIN study.
- Author
-
Sborov, Douglas W., Baljevic, Muhamed, Reeves, Brandi, Laubach, Jacob, Efebera, Yvonne A., Rodriguez, Cesar, Costa, Luciano J., Chari, Ajai, Silbermann, Rebecca, Holstein, Sarah A., Anderson, Larry D., Kaufman, Jonathan L., Shah, Nina, Pei, Huiling, Patel, Sharmila, Cortoos, Annelore, Bartlett, J. Blake, Vermeulen, Jessica, Lin, Thomas S., and Voorhees, Peter M.
- Subjects
MULTIPLE myeloma ,DARATUMUMAB ,MONOCLONAL gammopathies ,LENALIDOMIDE ,BORTEZOMIB ,STEM cell transplantation - Abstract
Summary: Patients with multiple myeloma are at increased risk of vascular thromboembolic events (VTEs). This post hoc analysis evaluated VTEs in the randomised phase 2 GRIFFIN study (ClinicalTrials.gov Identifier: NCT02874742) that investigated lenalidomide/bortezomib/dexamethasone (RVd) ± daratumumab (D). Patients with newly diagnosed multiple myeloma who were eligible for autologous stem cell transplantation (ASCT) received D‐RVd/RVd induction, high‐dose therapy and ASCT, D‐RVd/RVd consolidation and up to 2 years of lenalidomide maintenance therapy ± D. VTE prophylaxis was recommended (at least aspirin, ≥162 mg daily) in accordance with International Myeloma Working Group guidelines. In the safety population (D‐RVd, n = 99; RVd, n = 102), VTEs occurred in 10.1% of D‐RVd patients and 15.7% of RVd patients; grade 2–4 VTEs occurred in 9.1% and 14.7%, respectively. Median time to the first onset of VTE was longer for D‐RVd versus RVd patients (305 days vs 119 days). Anti‐thrombosis prophylaxis use was similar between arms (D‐RVd, 84.8% vs RVd, 83.3%); among patients with VTEs, prophylaxis use at time of first VTE onset was 60.0% for D‐RVd and 68.8% for RVd. In summary, the addition of daratumumab to RVd did not increase the incidence of VTEs, but the cumulative VTE incidence was relatively high in this cohort and anti‐thrombotic prophylaxis use was suboptimal. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
41. High Infiltration of CD68+/CD163− Macrophages Is an Adverse Prognostic Factor after Neoadjuvant Chemotherapy in Esophageal and Gastric Adenocarcinoma.
- Author
-
Svensson, Maria C., Svensson, Maja, Nodin, Björn, Borg, David, Hedner, Charlotta, Hjalmarsson, Claes, Leandersson, Karin, and Jirström, Karin
- Published
- 2022
- Full Text
- View/download PDF
42. Targeting CD38 for acute leukemia.
- Author
-
Xushu Zhong and Hongbing Ma
- Subjects
ACUTE leukemia ,CD38 antigen ,MULTIPLE myeloma ,HEMATOLOGIC malignancies ,BISPECIFIC antibodies - Abstract
Acute leukemia (AL) is a hematological malignancy, and the prognosis of most AL patients hasn’t improved significantly, particularly for relapsed or refractory (R/R) AL. Therefore, new treatments for R/R adult acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) are urgently necessary. Novel developments have been made in AL treatment, including target and immune therapies. CD38 is one of the targets due to its high expression in many hematological malignancies, including multiple myeloma, ALL and a subset of AML. Consequently, targeting CD38 therapies, including CD38 monoclonal antibodies (mAbs), bispecific antibodies, and CAR-T cell therapy, exhibit promising efficacy in treating multiple myeloma without significant toxicity and are being explored in other hematological malignancies and nonhematological diseases. Herein, this review focuses on targeting CD38 therapies in ALL and AML, which demonstrate sound antileukemic effects in acute leukemia and are expected to become effective treatment methods. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
43. Daratumumab, cyclophosphamide, bortezomib, and dexamethasone for multiple myeloma: final results of the LYRA study.
- Author
-
Yimer, Habte, Melear, Jason, Faber, Edward, Bensinger, William I., Burke, John M., Narang, Mohit, Stevens, Don, Gray, Kathleen S., Lutska, Yana, Bobba, Padma, Qi, Keqin, Hoehn, Daniela, Qi, Ming, Lin, Thomas S., and Rifkin, Robert M.
- Subjects
MULTIPLE myeloma ,DARATUMUMAB ,BORTEZOMIB ,STEM cell transplantation ,CYCLOPHOSPHAMIDE - Abstract
In the primary analysis of LYRA, daratumumab + cyclophosphamide/bortezomib/dexamethasone (DARA + CyBorD) was effective and well tolerated in newly diagnosed multiple myeloma (NDMM) and relapsed multiple myeloma (RMM). We report the final analysis of LYRA (median months of follow-up: NDMM, 35.7; RMM, 35.3) after all patients completed study therapy, were followed for 36 months, or discontinued. Patients received DARA + CyBorD induction, autologous stem cell transplant (if eligible), and 12 months of daratumumab maintenance. Eighty-seven NDMM patients enrolled, 39 underwent transplant, and 63 completed maintenance. Rates of complete response or better were 48.7% and 29.8% for NDMM transplant and NDMM non-transplant patients, respectively, and 36-month progression-free survival rates were 69.3% and 72.6%. Grade 3/4 treatment-emergent adverse events occurred in 61.6% of NDMM patients. Efficacy and safety data are also reported for the smaller RMM cohort (n = 14). DARA + CyBorD followed by daratumumab maintenance was well tolerated and achieved deep, durable responses in NDMM and RMM. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
44. FcγR-Mediated Trogocytosis 2.0: Revisiting History Gives Rise to a Unifying Hypothesis.
- Author
-
Lindorfer, Margaret A. and Taylor, Ronald P.
- Subjects
CANCER cells ,CELL receptors ,ENDOTHELIAL cells ,CLINICAL immunology ,B cells - Abstract
There is increasing interest in the clinical implications and immunology of trogocytosis, a process in which the receptors on acceptor cells remove and internalize cognate ligands from donor cells. We have reported that this phenomenon occurs in cancer immunotherapy, in which cells that express FcγR remove and internalize CD20 and bound mAbs from malignant B cells. This process can be generalized to include other reactions including the immune adherence phenomenon and antibody-induced immunosuppression. We discuss in detail FcγR-mediated trogocytosis and the evidence supporting a proposed predominant role for liver sinusoidal endothelial cells via the action of the inhibitory receptor FcγRIIb2. We describe experiments to test the validity of this hypothesis. The elucidation of the details of FcγR-mediated trogocytosis has the potential to allow for the development of novel therapies that can potentially block or enhance this reaction, depending upon whether the process leads to unfavorable or positive biological effects. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
45. Targeting CD38 with Daratumumab Plus Chemotherapy for Patients with Advanced-Stage Plasmablastoid Large B-Cell Lymphoma.
- Author
-
Ryu, Yun Kyoung, Ricker, Edd C., Soderquist, Craig R., Francescone, Mark A., Lipsky, Andrew H., and Amengual, Jennifer E.
- Subjects
DARATUMUMAB ,CD38 antigen ,LYMPHOMAS ,MULTIPLE myeloma ,MONOCLONAL antibodies - Abstract
Plasmablastic lymphoma (PBL) is a rare and aggressive form of large B-cell lymphoma (LBCL) most commonly seen in the setting of chronic immunosuppression or autoimmune disease. The prognosis is poor and CHOP-like regimens often fail to produce durable remission; therefore, there is no established standard of care treatment. However, PBL demonstrates substantial morphologic and immunophenotypic overlap with multiple myeloma (MM), suggesting that MM therapeutics might prove useful in treating PBL. We studied the effects of treatment using the first-in-class monoclonal antibody directed against CD38, daratumumab, in combination with chemotherapy in seven patients with advanced-stage LBCL with plasmablastic features. Treatment was safe and well-tolerated. Among six evaluable patients, six patients had complete response after treatment, and four patients who met strict WHO criteria for PBL had durable response (12–31 months and ongoing). [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
46. Daratumumab plus bortezomib or daratumumab plus lenalidomide as salvage therapy for patients with myeloma: initial follow-up of an Italian multicentre retrospective clinical experience by 'Rete Ematologica Pugliese'.
- Author
-
Mele, G., Cascavilla, N., Di Renzo, N., Guarini, A., Mazza, P., Melillo, L., Pavone, V., Tarantini, G., Curci, P., Falcone, A. P., Germano, C., Mele, A., Palazzo, G., Palumbo, G., Reddiconto, G., Rossini, B., Specchia, G., Musto, P., and Pastore, D.
- Abstract
We report herein a multicentre retrospective analysis of 192 consecutive patients with symptomatic refractory/relapsed multiple myeloma (RRMM) treated with daratumumab in combination with bortezomib or lenalidomide as salvage therapy at 9 haematological centres in Puglia. Choice of both regimens was based on previous treatment and/or physicians' preference. Considering the under-representation of older patients (very old patient ≥ 80 years) in clinical trials and the prognostic and predictive importance and value of frailty status, here, we further characterised the patient cohort by age. The overall response rate (ORR) was generally lower than what was previously reported in the CASTOR (ORR 72.6% vs 85%) and POLLUX (ORR 86.5% vs 93%) trials. The lower ORR in our analysis compared to the CASTOR and POLLUX trials could be related to a less selected population. Similarly, amongst very old patients, the ORR was encouraging: ORR to treatment with DVd (daratumumab + bortezomib + dexamethasone) was 66.7%, and ORR to treatment with DRd (daratumumab + lenalidomide + dexamethasone) was 92.3%. Median TTP (time to progression) was 10.8 months (1-year TTP: 44.7%; 2-year TTP: 25.3%) in the DVd group; median TTP was not reached in the DRd group (1-year TTP: 82.7%; 2-year TTP: 71.4%). Median OS (overall survival) was not reached either in the DRd group (1-year OS: 85.9%; 2-year OS: 73.7%) or the DVd group (1-year OS: 70.2%; 2-year OS: 58.9%). [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
47. Daratumumab for the Treatment of Multiple Myeloma: A Review of Clinical Applicability and Operational Considerations.
- Author
-
Arnall, Justin R., Maples, Kathryn T., Harvey, R. Donald, and Moore, Donald C.
- Abstract
Objective: To review the available data for the efficacy and safety of daratumumab in the treatment of multiple myeloma (MM), both in the newly diagnosed and relapsed/refractory settings, as well as provide additional guidance to clinicians on operational, safety, and supportive care considerations. Data Sources: A literature search of PubMed (1966 to October 2021) was conducted using the keywords daratumumab, Darzalex, and myeloma. Data were also obtained from prescribing information and unpublished abstracts from meetings. Study Selection and Data Extraction: All relevant published articles, prescribing information, and unpublished meeting abstracts on daratumumab for the treatment of MM were reviewed. Data Synthesis: Daratumumab is an anti-CD38 monoclonal antibody indicated for the treatment of MM. The addition of daratumumab to proteasome inhibitor and immunomodulatory drug-based regimens has led to a consistent improvement in progression-free survival and response rates in relapsed/refractory MM as per the POLLUX, CASTOR, APOLLO, and CANDOR trials. The ALCYONE and MAIA phase III trials have demonstrated an overall survival benefit when adding daratumumab to frontline regimens for transplant-ineligible patients with newly diagnosed MM. In transplant-eligible patients, daratumumab-based quadruplet regimens have improved depth of response in the CASSIOPIEA and GRIFFIN trials. Relevance to Patient Care and Clinical Practice: Operational and safety considerations that clinicians need to account for do exist, including different administration and infusion strategies, infusion-related reactions, increased risk for infectious complications, and interference with blood transfusion management. Conclusions: Daratumumab has led to a shift in the treatment paradigm of both newly diagnosed and relapsed/refractory MM, leading to improvements in outcomes such as response rates, depth of response, and progression-free survival. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
48. Development and Characterization of an Anti-Cancer Monoclonal Antibody for Treatment of Human Carcinomas.
- Author
-
Tsang, Kwong yok, Fantini, Massimo, Mavroukakis, Sharon A., Zaki, Anjum, Annunziata, Christina M., and Arlen, Philip M.
- Subjects
THERAPEUTIC use of monoclonal antibodies ,PROGRAMMED cell death 1 receptors ,CLINICAL drug trials ,IMMUNE checkpoint inhibitors ,COMPLEMENT (Immunology) ,KILLER cells ,CELLULAR signal transduction ,CELL adhesion molecules ,TUMORS ,DRUG development ,CELL lines ,IMMUNOTHERAPY ,PHARMACODYNAMICS - Abstract
Simple Summary: Cancers can grow and spread to different parts of the body. The aim of immunotherapy is to stimulate the immune system to eliminate cancer cells in a selective manner. Tumor-targeting monoclonal antibodies (mAb) can be used as immunotherapy to stimulate innate antitumor immunity. In this review, we have described the development and characterization of an anti-cancers mAb NEO-201. NEO-201 is an IgG1 humanized mAb that binds specifically to tumor-associated variants of CEACAM-5 and CEACAM-6 expressed by colon, ovarian, pancreatic, non-small cell lung, head and neck, cervical, uterine and breast cancers, but is not reactive against most normal tissues. The peculiarity of NEO-201 is its ability to counteract tumor growth through different mechanisms. NEO-201 engages components of immune system to kill tumor cells expressing its target via antibody-dependent cell-mediated cytotoxicity and complement dependent cytotoxicity. NEO-201 can also indirectly enhance anti-cancer activity through the blockade of the interaction between CEACAM-5 expressed on tumor cells and CEACAM-1 expressed on natural killer cells to reverse CEACAM-1-dependent inhibition of NK cytotoxicity or through its binding to human regulatory T cells. The specificity of NEO-201 in recognizing suppressive regulatory T cells provides the basis for combination cancer immunotherapy with checkpoint inhibitors targeting the PD-1/PD-L1 pathway. NEO-201 is an IgG1 humanized monoclonal antibody (mAb) that binds to tumor-associated variants of carcinoembryonic antigen-related cell adhesion molecule (CEACAM)-5 and CEACAM-6. NEO-201 reacts to colon, ovarian, pancreatic, non-small cell lung, head and neck, cervical, uterine and breast cancers, but is not reactive against most normal tissues. NEO-201 can kill tumor cells via antibody-dependent cell-mediated cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC) to directly kill tumor cells expressing its target. We explored indirect mechanisms of its action that may enhance immune tumor killing. NEO-201 can block the interaction between CEACAM-5 expressed on tumor cells and CEACAM-1 expressed on natural killer (NK) cells to reverse CEACAM-1-dependent inhibition of NK cytotoxicity. Previous studies have demonstrated safety/tolerability in non-human primates, and in a first in human phase 1 clinical trial at the National Cancer Institute (NCI). In addition, preclinical studies have demonstrated that NEO-201 can bind to human regulatory T (Treg) cells. The specificity of NEO-201 in recognizing suppressive Treg cells provides the basis for combination cancer immunotherapy with checkpoint inhibitors targeting the PD-1/PD-L1 pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
49. Multiplex Immunofluorescence and the Digital Image Analysis Workflow for Evaluation of the Tumor Immune Environment in Translational Research.
- Author
-
Rojas, Frank, Hernandez, Sharia, Lazcano, Rossana, Laberiano-Fernandez, Caddie, and Parra, Edwin Roger
- Subjects
IMAGE analysis ,TUMOR microenvironment ,TRANSLATIONAL research ,IMMUNOFLUORESCENCE - Abstract
A robust understanding of the tumor immune environment has important implications for cancer diagnosis, prognosis, research, and immunotherapy. Traditionally, immunohistochemistry (IHC) has been regarded as the standard method for detecting proteins in situ , but this technique allows for the evaluation of only one cell marker per tissue sample at a time. However, multiplexed imaging technologies enable the multiparametric analysis of a tissue section at the same time. Also, through the curation of specific antibody panels, these technologies enable researchers to study the cell subpopulations within a single immunological cell group. Thus, multiplexed imaging gives investigators the opportunity to better understand tumor cells, immune cells, and the interactions between them. In the multiplexed imaging technology workflow, once the protocol for a tumor immune micro environment study has been defined, histological slides are digitized to produce high-resolution images in which regions of interest are selected for the interrogation of simultaneously expressed immunomarkers (including those co-expressed by the same cell) by using an image analysis software and algorithm. Most currently available image analysis software packages use similar machine learning approaches in which tissue segmentation first defines the different components that make up the regions of interest and cell segmentation, then defines the different parameters, such as the nucleus and cytoplasm, that the software must utilize to segment single cells. Image analysis tools have driven dramatic evolution in the field of digital pathology over the past several decades and provided the data necessary for translational research and the discovery of new therapeutic targets. The next step in the growth of digital pathology is optimization and standardization of the different tasks in cancer research, including image analysis algorithm creation, to increase the amount of data generated and their accuracy in a short time as described herein. The aim of this review is to describe this process, including an image analysis algorithm creation for multiplex immunofluorescence analysis, as an essential part of the optimization and standardization of the different processes in cancer research, to increase the amount of data generated and their accuracy in a short time. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
50. Identification of New Antibodies Targeting Malignant Plasma Cells for Immunotherapy by Next-Generation Sequencing-Assisted Phage Display.
- Author
-
Krohn, Steffen, Boje, Ammelie Svea, Gehlert, Carina Lynn, Lutz, Sebastian, Darzentas, Nikos, Knecht, Henrik, Herrmann, Dietrich, Brüggemann, Monika, Scheidig, Axel J., Weisel, Katja, Gramatzki, Martin, Peipp, Matthias, and Klausz, Katja
- Subjects
PLASMA cells ,CANCER cells ,CHO cell ,MONONUCLEAR leukocytes ,IMMUNOGLOBULINS - Abstract
To identify new antibodies for the treatment of plasma cell disorders including multiple myeloma (MM), a single-chain Fragment variable (scFv) antibody library was generated by immunizing mice with patient-derived malignant plasma cells. To enrich antibodies binding myeloma antigens, phage display with cellular panning was performed. After depleting the immune library with leukocytes of healthy donors, selection of antibodies was done with L-363 plasma cell line in two consecutive panning rounds. Monitoring the antibodies' enrichment throughout the panning by next-generation sequencing (NGS) identified several promising candidates. Initially, 41 unique scFv antibodies evolving from different B cell clones were selected. Nine of these antibodies strongly binding to myeloma cells and weakly binding to peripheral blood mononuclear cells (PBMC) were characterized. Using stably transfected Chinese hamster ovary cells expressing individual myeloma-associated antigens revealed that two antibodies bind CD38 and intercellular adhesion molecule-1 (ICAM-1), respectively, and 7 antibodies target yet unknown antigens. To evaluate the therapeutic potential of our new antibodies, in a first proof-of-concept study the CD38 binding scFv phage antibody was converted into a chimeric IgG1. Further analyses revealed that #5-CD38-IgG1 shared an overlapping epitope with daratumumab and isatuximab and had potent anti-myeloma activity comparable to the two clinically approved CD38 antibodies. These results indicate that by phage display and deep sequencing, new antibodies with therapeutic potential for MM immunotherapy can be identified. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.