Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (51)

Search Parameters:
Keywords = immunomodulatory imide drugs

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 1135 KiB  
Article
Serum Cereblon (CRBN) Levels Predict Long Term Post- Lenalidomide-Dexamethasone Survival in Multiple Myeloma (MM) Patients and Correlate with Disease Characteristics
by Annita-Ioanna Gkioka, Alexandros Gkiokas, Mavra Papadatou-Gigante, Alexandros Alexandropoulos, Thomai-Marina Tryfou, Aspasia Koudouna, Vasiliki Bartzi and Marie-Christine Kyrtsonis
Int. J. Mol. Sci. 2025, 26(13), 6341; https://doi.org/10.3390/ijms26136341 - 30 Jun 2025
Viewed by 371
Abstract
Serum cereblon (CRBN) has been proposed as a target protein for immunomodulatory drugs (IMiDs). IMiDs are one of the backbone treatment options in multiple myeloma (MM), rendering CRBN an intriguing candidate for use as a biomarker in clinical settings. Ninety-two (92) MM patients, [...] Read more.
Serum cereblon (CRBN) has been proposed as a target protein for immunomodulatory drugs (IMiDs). IMiDs are one of the backbone treatment options in multiple myeloma (MM), rendering CRBN an intriguing candidate for use as a biomarker in clinical settings. Ninety-two (92) MM patients, mostly relapsed/refractory and a few at diagnosis, were included in the study, from lenalidomide–dexamethasone (LD) initiation until last follow-up or death. Median CRBN at LD initiation (N = 68) treatment was 247 pg/mL (range, 0–9760 pg/mL), at the time of best response (BR) status (N = 59) 142.5 pg/mL (range, 0–9940 pg/mL) and in patients with relapse/refractory MM to LD regimen (N = 54) 298 pg/mL (range, 0–9840 pg/mL). CRBN in healthy individuals was almost undetectable and significantly lower compared to the CRBN at LD initiation (p = 0.003), at BR to LD (p = 0.012) and at relapse to LD (p = 0.002). CRBN was significantly lower at BR in contrast to LD initiation and relapse to LD (p = 0.04, p = 0.028). High levels of CRBN at treatment initiation correlated with early relapse to LD (≤12 months) (p = 0.03). Seven-year survival was improved in patients with CRBN levels below median measured at the time of LD initiation (p = 0.013) as well as at BR (p = 0.032). CRBN was associated with treatment response and is predictive of survival after LD. Full article
(This article belongs to the Special Issue Immunomodulatory Molecules in Cancer)
Show Figures

Figure 1

16 pages, 1850 KiB  
Systematic Review
Upfront Anti-CD38 Monoclonal Antibody-Based Quadruplet Therapy for Multiple Myeloma: A Systematic Review and Meta-Analysis of Clinical Trials
by Ioannis Ntanasis-Stathopoulos, Charalampos Filippatos, Panagiotis Malandrakis, Vassilis Koutoulidis, Efstathios Kastritis, Evangelos Terpos, Meletios-Athanasios Dimopoulos and Maria Gavriatopoulou
Cancers 2025, 17(12), 1943; https://doi.org/10.3390/cancers17121943 - 11 Jun 2025
Viewed by 1031
Abstract
Background: Recently, the addition of anti-CD38 monoclonal antibodies (mAbs) to standard first-line triplet regimens, including a proteasome inhibitor (PI), an immunomodulatory drug (IMiD) and dexamethasone, has led to the introduction of quadruplets in clinical practice. Methods: A systematic search was conducted (end-of-search: 9 [...] Read more.
Background: Recently, the addition of anti-CD38 monoclonal antibodies (mAbs) to standard first-line triplet regimens, including a proteasome inhibitor (PI), an immunomodulatory drug (IMiD) and dexamethasone, has led to the introduction of quadruplets in clinical practice. Methods: A systematic search was conducted (end-of-search: 9 November, 2024) for clinical trials investigating first-line anti-CD38 mAb-based quadruplets in combination with a PI and an IMiD. Pooled proportions and effect-estimates along with 95% confidence intervals were calculated with common-effect and random-effects models and further subgroup and meta-regression analyses were performed. Results: The pooled 2-, 3- and 4-year progression-free survival (PFS) rates were 89%, 77% and 86%, respectively. Furthermore, patients treated with quadruplets demonstrated a 46% reduced risk for disease progression or death (HR = 0.54, 95% CI: 0.46–0.64) compared to those on triplets. Overall survival (OS) rates were consistently high, ranging from 83% to 96% between different regimens. High rates of deep responses that deepened over time were observed, with the pooled proportion of patients achieving at least complete response being 64%. Importantly, the pooled MRD negativity rate was 62%, whereas patients treated with quadruplet first-line therapy had 2.5 times the odds to be MRD negative at any point compared with those on triplets. Moreover, the odds for sustained 12-month MRD negativity were thrice as much with quadruplets compared to triplets. Finally, while no increase in serious adverse events was observed with quadruplet regimens compared to triplets, a 46% statistically significant increased risk for grade 3–4 neutropenia and thrombocytopenia was observed, along with a 14% increased risk for grade 3–4 infections. Conclusions: The addition of anti-CD38 mAbs to standard triplet regimens has shown particularly favorable outcomes, supporting their integration in the upfront treatment of patients with NDMM. However, close monitoring for hematological toxicity and infections is essential. Full article
Show Figures

Figure 1

36 pages, 3058 KiB  
Review
Post-Translational Modifications in Multiple Myeloma: Mechanisms of Drug Resistance and Therapeutic Opportunities
by Shuoyang Hu, Jirun Xu, Weiyan Cui, Haoran Jin, Xiaoyu Wang and Yasen Maimaitiyiming
Biomolecules 2025, 15(5), 702; https://doi.org/10.3390/biom15050702 - 12 May 2025
Viewed by 957
Abstract
Multiple myeloma (MM) remains an incurable hematologic malignancy due to the inevitable development of drug resistance, particularly in relapsed or refractory cases. Post-translational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, and glycosylation, play pivotal roles in regulating protein function, stability, and interactions, thereby influencing [...] Read more.
Multiple myeloma (MM) remains an incurable hematologic malignancy due to the inevitable development of drug resistance, particularly in relapsed or refractory cases. Post-translational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, and glycosylation, play pivotal roles in regulating protein function, stability, and interactions, thereby influencing MM pathogenesis and therapeutic resistance. This review comprehensively explores the mechanisms by which dysregulated PTMs contribute to drug resistance in MM, focusing on their impact on key signaling pathways, metabolic reprogramming, and the tumor microenvironment. We highlight how PTMs modulate drug uptake, alter drug targets, and regulate cell survival signals, ultimately promoting resistance to PIs, IMiDs, and other therapeutic agents. Furthermore, we discuss emerging therapeutic strategies targeting PTM-related pathways, which offer promising avenues for overcoming resistance to treatment. By integrating preclinical and clinical insights, this review underscores the potential of PTM-targeted therapies to enhance treatment efficacy and improve patient outcomes in MM. Full article
(This article belongs to the Section Cellular Biochemistry)
Show Figures

Figure 1

11 pages, 1657 KiB  
Article
Evolution of Survival in Patients with Multiple Myeloma over Two Decades: A Real-World Experience from a Medium-Level Hospital
by Esther Ortega-Vida, Abel Rosado-Rodriguez, Rocio Fe, Victoria Verdugo, Rocio Gavira and Sebastián Garzón
Cancers 2025, 17(5), 793; https://doi.org/10.3390/cancers17050793 - 25 Feb 2025
Cited by 1 | Viewed by 1156
Abstract
Background: The gradual introduction of numerous therapeutic advancements over recent years in the treatment of patients with multiple myeloma (MM) appears to have contributed to significant improvements in overall survival (OS). Methods: We conducted a single-center retrospective observational study, including all MM patients [...] Read more.
Background: The gradual introduction of numerous therapeutic advancements over recent years in the treatment of patients with multiple myeloma (MM) appears to have contributed to significant improvements in overall survival (OS). Methods: We conducted a single-center retrospective observational study, including all MM patients treated at the University Hospital of Jerez de la Frontera, diagnosed between 1 January 2000, and 31 December 2022. Patients were divided into three calendar periods (2000–2007, 2008–2015, and 2016–2022) and two patient groups (candidates and non-candidates for autologous hematopoietic progenitor transplantation). Results: A total of 420 myeloma patients were included in this study, with a median age of 64 years. The median survival steadily improved from 50.7 months (33.8–73.2; 95% CI) in 2000–2007 to 72.4 months (57.5–98.2; 95% CI) in 2008–2015 and has not yet been determined in the 2016–2022 cohort (p = 0.008). OS improved in all age groups, even in older patients. The median OS in patients not undergoing autologous stem cell transplantation (ASCT) was 38.8 months (31.5–51.6; 95% CI) compared with 132.66 months (110.5–150.9; 95% CI) in those who underwent this procedure (p < 0.0001). The introduction of novel drug classes in first-line treatment significantly improved OS compared with traditional chemotherapy (56.7 months). The median OS increased to 78.8 months (95% CI: 68.8–113.3; p = 0.03) with proteasome inhibitors (PIs), 99.4 months (95% CI: 99.4–NA; < 0.0001) with immunomodulatory drugs (IMIDs), and was not determined for anti-CD38 monoclonal antibodies (p = 0.02). Conclusions: Survival outcomes for MM have significantly improved over the past two decades, particularly among younger and ASCT-eligible patients. However, according to all studies, disparities persist across healthcare settings, underscoring the need for equitable access to modern therapies and optimized management strategies. Full article
(This article belongs to the Section Cancer Survivorship and Quality of Life)
Show Figures

Figure 1

14 pages, 3283 KiB  
Article
Preclinical Evaluation of a Novel Series of Polyfluorinated Thalidomide Analogs in Drug-Resistant Multiple Myeloma
by Blaire E. Barton, Matthew K. Collins, Cindy H. Chau, Hyoyoung Choo-Wosoba, David J. Venzon, Christian Steinebach, Kathleen M. Garchitorena, Bhruga Shah, Eric L. Sarin, Michael Gütschow and William D. Figg
Biomolecules 2024, 14(6), 725; https://doi.org/10.3390/biom14060725 - 19 Jun 2024
Cited by 1 | Viewed by 1998
Abstract
Immunomodulatory imide drugs (IMiDs) play a crucial role in the treatment landscape across various stages of multiple myeloma. Despite their evident efficacy, some patients may exhibit primary resistance to IMiD therapy, and acquired resistance commonly arises over time leading to inevitable relapse. It [...] Read more.
Immunomodulatory imide drugs (IMiDs) play a crucial role in the treatment landscape across various stages of multiple myeloma. Despite their evident efficacy, some patients may exhibit primary resistance to IMiD therapy, and acquired resistance commonly arises over time leading to inevitable relapse. It is critical to develop novel therapeutic options to add to the treatment arsenal to overcome IMiD resistance. We designed, synthesized, and screened a new class of polyfluorinated thalidomide analogs and investigated their anti-cancer, anti-angiogenic, and anti-inflammatory activity using in vitro and ex vivo biological assays. We identified four lead compounds that exhibit potent anti-myeloma, anti-angiogenic, anti-inflammatory properties using three-dimensional tumor spheroid models, in vitro tube formation, and ex vivo human saphenous vein angiogenesis assays, as well as the THP-1 inflammatory assay. Western blot analyses investigating the expression of proteins downstream of cereblon (CRBN) reveal that Gu1215, our primary lead candidate, exerts its activity through a CRBN-independent mechanism. Our findings demonstrate that the lead compound Gu1215 is a promising candidate for further preclinical development to overcome intrinsic and acquired IMiD resistance in multiple myeloma. Full article
Show Figures

Figure 1

15 pages, 888 KiB  
Review
IMiD-Free Interval and IMiDs Sequence: Which Strategy Is Better Suited for Lenalidomide-Refractory Myeloma?
by Kazuhito Suzuki and Shingo Yano
Life 2023, 13(11), 2229; https://doi.org/10.3390/life13112229 - 20 Nov 2023
Cited by 1 | Viewed by 2460
Abstract
This review discusses immunomodulatory drug (IMiDs) sequencing and IMiD-free interval strategies for lenalidomide-refractory myeloma. IMiDs and proteasome inhibitors (PIs) improve clinical outcomes in patients with myeloma; however, refractoriness to lenalidomide, a category of IMiD, predicts poor outcomes. Next-generation IMiDs, such as pomalidomide, are [...] Read more.
This review discusses immunomodulatory drug (IMiDs) sequencing and IMiD-free interval strategies for lenalidomide-refractory myeloma. IMiDs and proteasome inhibitors (PIs) improve clinical outcomes in patients with myeloma; however, refractoriness to lenalidomide, a category of IMiD, predicts poor outcomes. Next-generation IMiDs, such as pomalidomide, are effective even for lenalidomide-refractory myeloma. Therefore, an IMiD-sequencing strategy from lenalidomide to pomalidomide would be desirable. PIs are an antimyeloma therapeutic agent with another mode of action that might restore cereblon, a target of IMiDs; therefore, an IMiD-free interval via class switching from lenalidomide to PIs may be a promising alternative for lenalidomide-refractory myeloma. Additionally, the anti-CD38 monoclonal antibody is a key drug for salvage therapy in anti-CD38 monoclonal antibody-naïve patients. In clinical practice, safety profiles and social convenience can play important roles in the choice of combination therapy. In the future, the selection of optimal treatments should be based on the status of the immunological environment and genetic alterations. This review aims to discuss IMiDs sequencing and IMiD-free interval strategies for lenalidomide- refractory myeloma. Full article
(This article belongs to the Special Issue Multiple Myeloma: Innovations in Diagnosis and Treatment)
Show Figures

Figure 1

9 pages, 563 KiB  
Communication
Clinical Outcomes of Patients with Multiple Myeloma after Daratumumab Failure
by Irene Zamanillo, Lucia Medina de Alba, Rodrigo Gil, Rosalia de la Puerta, Rafael Alonso, Ana Jimenez-Ubieto, Maria Teresa Cedena, Maria Calbacho, Rosa Ayala and Joaquin Martinez-Lopez
Life 2023, 13(9), 1841; https://doi.org/10.3390/life13091841 - 31 Aug 2023
Viewed by 1942
Abstract
Anti-CD38 monoclonal antibody (MoAB) therapy has significantly improved the prognosis of patients with multiple myeloma. However, not all patients sustain durable responses. We aimed to describe the natural history of patients relapsed or refractory (R/R) to CD38 MoAB therapy. We performed a single-center, [...] Read more.
Anti-CD38 monoclonal antibody (MoAB) therapy has significantly improved the prognosis of patients with multiple myeloma. However, not all patients sustain durable responses. We aimed to describe the natural history of patients relapsed or refractory (R/R) to CD38 MoAB therapy. We performed a single-center, retrospective analysis of the clinical characteristics and outcomes of 81 patients with multiple myeloma who progressed after treatment with daratumumab. Our cohort was heavily pretreated, with a median of two lines prior to daratumumab and only 17 patients received daratumumab as a first line. A total of 38.2% had received a previous autologous stem cell transplantation (ASCT), and 61.7% had received both an immunomodulatory drug (IMID) and a proteasome inhibitor (PI). The median overall survival (OS) was 21 months for the global cohort but it decreased to 14 months for triple-class refractory patients and 5 months for penta-refractory patients. Most of the patients (83.9%) received treatment after daratumumab progression, in many cases with second generation IMID or PI, but seven patients were treated with anti-BCMA therapy and three patients received CART therapy within a clinical trial. In conclusion, patients R/R to daratumumab represent an unmet clinical need with poor prognosis and in need of incorporation of new treatments. Full article
Show Figures

Figure 1

19 pages, 2377 KiB  
Article
A New Strategy for the Old Challenge of Thalidomide: Systems Biology Prioritization of Potential Immunomodulatory Drug (IMiD)-Targeted Transcription Factors
by Thayne Woycinck Kowalski, Mariléa Furtado Feira, Vinícius Oliveira Lord, Julia do Amaral Gomes, Giovanna Câmara Giudicelli, Lucas Rosa Fraga, Maria Teresa Vieira Sanseverino, Mariana Recamonde-Mendoza, Lavinia Schuler-Faccini and Fernanda Sales Luiz Vianna
Int. J. Mol. Sci. 2023, 24(14), 11515; https://doi.org/10.3390/ijms241411515 - 15 Jul 2023
Cited by 2 | Viewed by 3090
Abstract
Several molecular mechanisms of thalidomide embryopathy (TE) have been investigated, from anti-angiogenesis to oxidative stress to cereblon binding. Recently, it was discovered that thalidomide and its analogs, named immunomodulatory drugs (IMiDs), induced the degradation of C2H2 transcription factors (TFs). This mechanism might impact [...] Read more.
Several molecular mechanisms of thalidomide embryopathy (TE) have been investigated, from anti-angiogenesis to oxidative stress to cereblon binding. Recently, it was discovered that thalidomide and its analogs, named immunomodulatory drugs (IMiDs), induced the degradation of C2H2 transcription factors (TFs). This mechanism might impact the strict transcriptional regulation of the developing embryo. Hence, this study aims to evaluate the TFs altered by IMiDs, prioritizing the ones associated with embryogenesis through transcriptome and systems biology-allied analyses. This study comprises only the experimental data accessed through bioinformatics databases. First, proteins and genes reported in the literature as altered/affected by the IMiDs were annotated. A protein systems biology network was evaluated. TFs beta-catenin (CTNNB1) and SP1 play more central roles: beta-catenin is an essential protein in the network, while SP1 is a putative C2H2 candidate for IMiD-induced degradation. Separately, the differential expressions of the annotated genes were analyzed through 23 publicly available transcriptomes, presenting 8624 differentially expressed genes (2947 in two or more datasets). Seventeen C2H2 TFs were identified as related to embryonic development but not studied for IMiD exposure; these TFs are potential IMiDs degradation neosubstrates. This is the first study to suggest an integration of IMiD molecular mechanisms through C2H2 TF degradation. Full article
Show Figures

Figure 1

34 pages, 6792 KiB  
Review
A New Generation of IMiDs as Treatments for Neuroinflammatory and Neurodegenerative Disorders
by Katherine O. Kopp, Margaret E. Greer, Elliot J. Glotfelty, Shih-Chang Hsueh, David Tweedie, Dong Seok Kim, Marcella Reale, Neil Vargesson and Nigel H. Greig
Biomolecules 2023, 13(5), 747; https://doi.org/10.3390/biom13050747 - 26 Apr 2023
Cited by 13 | Viewed by 5856
Abstract
The immunomodulatory imide drug (IMiD) class, which includes the founding drug member thalidomide and later generation drugs, lenalidomide and pomalidomide, has dramatically improved the clinical treatment of specific cancers, such as multiple myeloma, and it combines potent anticancer and anti-inflammatory actions. These actions, [...] Read more.
The immunomodulatory imide drug (IMiD) class, which includes the founding drug member thalidomide and later generation drugs, lenalidomide and pomalidomide, has dramatically improved the clinical treatment of specific cancers, such as multiple myeloma, and it combines potent anticancer and anti-inflammatory actions. These actions, in large part, are mediated by IMiD binding to the human protein cereblon that forms a critical component of the E3 ubiquitin ligase complex. This complex ubiquitinates and thereby regulates the levels of multiple endogenous proteins. However, IMiD-cereblon binding modifies cereblon’s normal targeted protein degradation towards a new set of neosubstrates that underlies the favorable pharmacological action of classical IMiDs, but also their adverse actions—in particular, their teratogenicity. The ability of classical IMiDs to reduce the synthesis of key proinflammatory cytokines, especially TNF-α levels, makes them potentially valuable to reposition as drugs to mitigate inflammatory-associated conditions and, particularly, neurological disorders driven by an excessive neuroinflammatory element, as occurs in traumatic brain injury, Alzheimer’s and Parkinson’s diseases, and ischemic stroke. The teratogenic and anticancer actions of classical IMiDs are substantial liabilities for effective drugs in these disorders and can theoretically be dialed out of the drug class. We review a select series of novel IMiDs designed to avoid binding with human cereblon and/or evade degradation of downstream neosubstrates considered to underpin the adverse actions of thalidomide-like drugs. These novel non-classical IMiDs hold potential as new medications for erythema nodosum leprosum (ENL), a painful inflammatory skin condition associated with Hansen’s disease for which thalidomide remains widely used, and, in particular, as a new treatment strategy for neurodegenerative disorders in which neuroinflammation is a key component. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Graphical abstract

17 pages, 1111 KiB  
Review
Bispecific Antibodies in Multiple Myeloma: Opportunities to Enhance Efficacy and Improve Safety
by Dawn Swan, Philip Murphy, Siobhan Glavey and John Quinn
Cancers 2023, 15(6), 1819; https://doi.org/10.3390/cancers15061819 - 17 Mar 2023
Cited by 27 | Viewed by 7924
Abstract
Multiple myeloma (MM) is the second most common haematological neoplasm of adults in the Western world. Overall survival has doubled since the advent of proteosome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies. However, patients with adverse cytogenetics or high-risk disease as determined [...] Read more.
Multiple myeloma (MM) is the second most common haematological neoplasm of adults in the Western world. Overall survival has doubled since the advent of proteosome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies. However, patients with adverse cytogenetics or high-risk disease as determined by the Revised International Staging System (R-ISS) continue to have poorer outcomes, and triple-refractory patients have a median survival of less than 1 year. Bispecific antibodies (BsAbs) commonly bind to a tumour epitope along with CD3 on T-cells, leading to T-cell activation and tumour cell killing. These treatments show great promise in MM patients, with the first agent, teclistamab, receiving regulatory approval in 2022. Their potential utility is hampered by the immunosuppressive tumour microenvironment (TME), a hallmark of MM, which may limit efficacy, and by undesirable adverse events, including cytokine release syndrome (CRS) and infections, some of which may be fatal. In this review, we first consider the means of enhancing the efficacy of BsAbs in MM. These include combining BsAbs with other drugs that ameliorate the effect of the immunosuppressive TME, improving target availability, the use of BsAbs directed against multiple target antigens, and the optimal time in the treatment pathway to employ BsAbs. We then discuss methods to improve safety, focusing on reducing infection rates associated with treatment-induced hypogammaglobulinaemia, and decreasing the frequency and severity of CRS. BsAbs offer a highly-active therapeutic option in MM. Improving the efficacy and safety profiles of these agents may enable more patients to benefit from these novel therapies and improve outcomes for patients with high-risk disease. Full article
Show Figures

Figure 1

26 pages, 899 KiB  
Review
Relapsed/Refractory Multiple Myeloma: A Review of Available Therapies and Clinical Scenarios Encountered in Myeloma Relapse
by Parva Bhatt, Colin Kloock and Raymond Comenzo
Curr. Oncol. 2023, 30(2), 2322-2347; https://doi.org/10.3390/curroncol30020179 - 15 Feb 2023
Cited by 50 | Viewed by 12225
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 [...] Read more.
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). Full article
Show Figures

Figure 1

19 pages, 1718 KiB  
Review
Lenalidomide in Multiple Myeloma: Review of Resistance Mechanisms, Current Treatment Strategies and Future Perspectives
by Piotr Kulig, Sławomir Milczarek, Estera Bakinowska, Laura Szalewska, Bartłomiej Baumert and Bogusław Machaliński
Cancers 2023, 15(3), 963; https://doi.org/10.3390/cancers15030963 - 2 Feb 2023
Cited by 16 | Viewed by 7325
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy, accounting for approximately 1% of all cancers. Despite the initial poor prognosis for MM patients, their life expectancy has improved significantly with the development of novel agents. Immunomodulatory drugs (IMiDs) are widely used [...] Read more.
Multiple myeloma (MM) is the second most common hematologic malignancy, accounting for approximately 1% of all cancers. Despite the initial poor prognosis for MM patients, their life expectancy has improved significantly with the development of novel agents. Immunomodulatory drugs (IMiDs) are widely used in MM therapy. Their implementation has been a milestone in improving the clinical outcomes of patients. The first molecule belonging to the IMiDs was thalidomide. Subsequently, its novel derivatives, lenalidomide (LEN) and pomalidomide (POM), were implemented. Almost all MM patients are exposed to LEN, which is the most commonly used IMiD. Despite the potent anti-MM activity of LEN, some patients eventually relapse and become LEN-resistant. Drug resistance is one of the greatest challenges of modern oncology and has become the main cause of cancer treatment failures. The number of patients receiving LEN is increasing, hence the problem of LEN resistance has become a great obstacle for hematologists worldwide. In this review, we intended to shed more light on the pathophysiology of LEN resistance in MM, with particular emphasis on the molecular background. Moreover, we have briefly summarized strategies to overcome LEN resistance and we have outlined future directions. Full article
(This article belongs to the Special Issue Immunomodulatory Agents for Multiple Myeloma)
Show Figures

Figure 1

28 pages, 5612 KiB  
Article
Activity of a Novel Anti-Inflammatory Agent F-3,6′-dithiopomalidomide as a Treatment for Traumatic Brain Injury
by Shih Chang Hsueh, Michael T. Scerba, David Tweedie, Daniela Lecca, Dong Seok Kim, Abdul Mannan Baig, Yu Kyung Kim, Inho Hwang, Sun Kim, Warren R. Selman, Barry J. Hoffer and Nigel H. Greig
Biomedicines 2022, 10(10), 2449; https://doi.org/10.3390/biomedicines10102449 - 30 Sep 2022
Cited by 7 | Viewed by 4260
Abstract
Traumatic brain injury (TBI) is a major risk factor for several neurodegenerative disorders, including Parkinson’s disease (PD) and Alzheimer’s disease (AD). Neuroinflammation is a cause of later secondary cell death following TBI, has the potential to aggravate the initial impact, and provides a [...] Read more.
Traumatic brain injury (TBI) is a major risk factor for several neurodegenerative disorders, including Parkinson’s disease (PD) and Alzheimer’s disease (AD). Neuroinflammation is a cause of later secondary cell death following TBI, has the potential to aggravate the initial impact, and provides a therapeutic target, albeit that has failed to translate into clinical trial success. Thalidomide-like compounds have neuroinflammation reduction properties across cellular and animal models of TBI and neurodegenerative disorders. They lower the generation of proinflammatory cytokines, particularly TNF-α which is pivotal in microglial cell activation. Unfortunately, thalidomide-like drugs possess adverse effects in humans before achieving anti-inflammatory drug levels. We developed F-3,6′-dithiopomalidomide (F-3,6′-DP) as a novel thalidomide-like compound to ameliorate inflammation. F-3,6′-DP binds to cereblon but does not efficiently trigger the degradation of the transcription factors (SALL4, Ikaros, and Aiolos) associated with the teratogenic and anti-proliferative responses of thalidomide-like drugs. We utilized a phenotypic drug discovery approach that employed cellular and animal models in the selection and development of F-3,6’-DP. F-3,6′-DP significantly mitigated LPS-induced inflammatory markers in RAW 264.7 cells, and lowered proinflammatory cytokine/chemokine levels in the plasma and brain of rats challenged with systemic LPS. We subsequently examined immunohistochemical, biochemical, and behavioral measures following controlled cortical impact (CCI) in mice, a model of moderate TBI known to induce inflammation. F-3,6′-DP decreased CCI-induced neuroinflammation, neuronal loss, and behavioral deficits when administered after TBI. F-3,6′-DP represents a novel class of thalidomide-like drugs that do not lower classical cereblon-associated transcription factors but retain anti-inflammatory actions and possess efficacy in the treatment of TBI and potentially longer-term neurodegenerative disorders. Full article
(This article belongs to the Special Issue Immune Dysfunction in Brain Disorders)
Show Figures

Figure 1

16 pages, 1351 KiB  
Review
CRL4CRBN E3 Ligase Complex as a Therapeutic Target in Multiple Myeloma
by Joanna Barankiewicz, Aleksander Salomon-Perzyński, Irena Misiewicz-Krzemińska and Ewa Lech-Marańda
Cancers 2022, 14(18), 4492; https://doi.org/10.3390/cancers14184492 - 16 Sep 2022
Cited by 22 | Viewed by 6484
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy with a recurrent clinical course. The introduction of immunomodulatory drugs (IMiDs) was one of the milestones in MM therapy leading to a significant improvement in patients’ prognosis. Currently, IMiDs are the backbone of [...] Read more.
Multiple myeloma (MM) is the second most common hematological malignancy with a recurrent clinical course. The introduction of immunomodulatory drugs (IMiDs) was one of the milestones in MM therapy leading to a significant improvement in patients’ prognosis. Currently, IMiDs are the backbone of MM therapy in newly diagnosed and relapsed/refractory settings. It is now known that IMiDs exert their anti-myeloma activity mainly by binding cereblon (CRBN), the substrate receptor protein of the CRL4 E3 ubiquitin ligase (CRL4CRBN) complex. By binding CRBN, IMiDs alter its substrate specificity, leading to ubiquitination and proteasomal degradation of proteins essential for MM cell survival. Following the success of IMiDs, it is not surprising that the possibility of using the CRL4CRBN complex’s activity to treat MM is being further explored. In this review, we summarize the current state of knowledge about novel players in the MM therapeutic landscape, namely the CRBN E3 ligase modulators (CELMoDs), the next generation of IMiDs with broader biological activity. In addition, we discuss a new strategy of tailored proteolysis called proteolysis targeting chimeras (PROTACs) using the CRL4CRBN to degrade typically undruggable proteins, which may have relevance for the treatment of MM and other malignancies in the future. Full article
(This article belongs to the Special Issue Immunomodulatory Agents for Multiple Myeloma)
Show Figures

Figure 1

9 pages, 262 KiB  
Review
Risk Factors and Risk Stratification of Thromboembolic Risk in Patients with Multiple Myeloma
by Roza Chaireti and Hareth Nahi
Hemato 2022, 3(3), 518-526; https://doi.org/10.3390/hemato3030036 - 29 Aug 2022
Viewed by 2377
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by a high risk for thrombotic episodes, mainly venous thromboembolism (VTE). This risk is accentuated by cancer treatments such as immunomodulatory drugs (IMiDs). Cancer-associated thrombosis is one of the leading causes of mortality and morbidity, [...] Read more.
Multiple myeloma (MM) is a hematological malignancy characterized by a high risk for thrombotic episodes, mainly venous thromboembolism (VTE). This risk is accentuated by cancer treatments such as immunomodulatory drugs (IMiDs). Cancer-associated thrombosis is one of the leading causes of mortality and morbidity, and the prevention of thrombosis is, therefore, of paramount significance. To this day, it is unclear which type of thromboprophylaxis is the most effective. This is partly due to the multifactorial etiology behind thrombosis since the compound of patient-, disease- and treatment-associated factors characterizing each patient with MM is unique. Additionally, the established risk scores are not reliable in patients with MM. The scope of this review is to summarize the factors contributing to increased thrombosis risk in MM, as well as the risk scores and thromboprophylaxis regimes available. Full article
(This article belongs to the Section Plasma Cell Disorders)
Back to TopTop