1
|
Pinney J, Roufosse C, Kousios A, Chaidos A, Gillmore JD, Rainone F, Choudhuri S, Ramasamy K, Blakey S, Ashcroft J, Chan YLT, Cockwell P, Pratt G. Diagnosis and management of monoclonal gammopathy of renal significance: A British Society for Haematology good practice paper. Br J Haematol 2025; 206:447-463. [PMID: 39777620 DOI: 10.1111/bjh.19956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025]
Abstract
This guideline provides consensus opinion on the investigations required for people presenting with suspected monoclonal gammopathy of renal significance to both nephrology and haematology physicians. The guideline discusses the principles of treating a patient with MGRS and provides recommendations for both supportive management and haematological therapy. It details the recommended on-going monitoring required for both specialty areas.
Collapse
Affiliation(s)
- Jennifer Pinney
- Department of Renal Medicine, University Hospital Birmingham NHS Trust, Birmingham, UK
| | - Candice Roufosse
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Andreas Kousios
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Aristeidis Chaidos
- Department of Immunology and Inflammation, Hugh and Josseline Langmuir Centre for Myeloma Research, Centre for Haematology, Imperial College London and Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Julian D Gillmore
- UK National Amyloidosis Centre, University College London, London, UK
| | - Francesco Rainone
- The Northern Care Alliance NHS Foundation Trust, Greater Manchester, UK
| | | | - Karthik Ramasamy
- Oxford University Hospital NHS Trust, and Oxford Translational Myeloma Centre, NDORMS, University of Oxford, Oxford, UK
| | - Sarah Blakey
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - John Ashcroft
- Department of Haematology, Mid Yorkshire Teaching Trust, Wakefield, UK
| | - Y L Tracey Chan
- Department of Renal Medicine, University Hospital Birmingham NHS Trust, Birmingham, UK
| | - Paul Cockwell
- Department of Renal Medicine, University Hospital Birmingham NHS Trust, Birmingham, UK
| | - Guy Pratt
- Department of Renal Medicine, University Hospital Birmingham NHS Trust, Birmingham, UK
| |
Collapse
|
2
|
Huynh T, Rodriguez-Rodriguez S, Danilov AV. Bruton Tyrosine Kinase Degraders in B-Cell Malignancies. Mol Cancer Ther 2024; 23:619-626. [PMID: 38693903 DOI: 10.1158/1535-7163.mct-23-0520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/02/2024] [Accepted: 02/29/2024] [Indexed: 05/03/2024]
Affiliation(s)
- Tiana Huynh
- City of Hope National Medical Center, Duarte, California
| | | | | |
Collapse
|
3
|
Niemann CU, Munir T, Moreno C, Owen C, Follows GA, Benjamini O, Janssens A, Levin MD, Robak T, Simkovic M, Voloshin S, Vorobyev V, Yagci M, Ysebaert L, Qi K, Qi Q, Sinet P, Parisi L, Srinivasan S, Schuier N, Baeten K, Howes A, Caces DB, Kater AP. Fixed-duration ibrutinib-venetoclax versus chlorambucil-obinutuzumab in previously untreated chronic lymphocytic leukaemia (GLOW): 4-year follow-up from a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2023; 24:1423-1433. [PMID: 37944541 DOI: 10.1016/s1470-2045(23)00452-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/24/2023] [Accepted: 09/06/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND In the GLOW study, fixed-duration ibrutinib-venetoclax showed superior progression-free survival versus chlorambucil-obinutuzumab in patients with previously untreated chronic lymphocytic leukaemia who were older or had comorbidities, or both, at a median follow up of 27·7 months. In this Article, we report updated outcomes from GLOW after a 46-month median follow-up. METHODS GLOW was a randomised, multicentre, phase 3 study done at 67 hospital centres across 14 countries. Patients aged 65 years and older or 18-64 years with previously untreated chronic lymphocytic leukaemia and a cumulative illness rating scale score of more than 6 or creatinine clearance less than 70 mL/min, or both, and an Eastern Cooperative Oncology Group performance status of 2 or less were randomly assigned (1:1) via an interactive web system with permuted blocks (block size of four) and stratified by IGHV mutational status and the presence of del11q aberration to the ibrutinib-venetoclax group (three cycles of ibrutinib lead-in [420 mg/day, orally], followed by 12 cycles of ibrutinib plus venetoclax [400 mg/day, orally, including a 5-week dose ramp-up]) or the chlorambucil-obinutuzumab group (six cycles of chlorambucil [0·5 mg/kg, orally, on days 1 and 15 of each cycle], and obinutuzumab [1000 mg, intravenously, on days 1 (or 100 mg on day 1 and 900 mg on day 2), 8, and 15 of cycle 1 and day 1 of cycles 2-6]). The primary endpoint was progression-free survival in the intention-to-treat population, assessed by an independent review committee. The safety population included all randomised patients who received at least one dose of the study treatment. This study is registered with ClinicalTrials.gov (NCT03462719) and the EU Clinical Trials Register (EudraCT 2017-004699-77). FINDINGS Between May 4, 2018, and April 5, 2019, 211 patients (122 [58%] were male and 89 [42%] were female) were randomly assigned to receive ibrutinib-venetoclax (n=106) or chlorambucil-obinutuzumab (n=105). At a median of 46 months (IQR 43-47) of follow-up, progression-free survival remained superior for the ibrutinib-venetoclax group (hazard ratio 0·214 [95% CI 0·138-0·334]; p<0·0001); 42-month progression-free survival rates were 74·6% (95% CI 65·0-82·0) for ibrutinib-venetoclax and 24·8% (16·5-34·1) for chlorambucil-obinutuzumab. Following the primary analysis, one patient in the chlorambucil-obinutuzumab group had a serious adverse event of myelodysplastic syndrome. Treatment-related deaths were reported in one patient receiving ibrutinib-venetoclax (cardiac failure, pneumonia, and sinus node dysfunction) and in one patient receiving chlorambucil-obinutuzumab (pneumonia). There were 15 deaths in the ibrutinib-venetoclax group (of which three were due to post-treatment infections) and 30 deaths in the chlorambucil-obinutuzumab group (of which 10 were due to post-treatment infections). INTERPRETATION After 4 years of follow-up, ibrutinib-venetoclax continues to significantly prolong progression-free survival (vs chemoimmunotherapy) in patients with previously untreated chronic lymphocytic leukaemia, supporting its use as a first-line option. FUNDING Janssen Research & Development and Pharmacyclics.
Collapse
Affiliation(s)
- Carsten U Niemann
- Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| | | | - Carol Moreno
- Hospital de la Santa Creu i Sant Pau, Autonomous University of Barcelona, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | | | | | | | | | | | - Tadeusz Robak
- Medical University of Lodz, Copernicus Memorial Hospital, Lodz, Poland
| | - Martin Simkovic
- 4th Department of Internal Medicine-Haematology, Faculty of Medicine in Hradec Králové, University Hospital and Charles University in Prague, Hradec Králové, Czech Republic
| | - Sergey Voloshin
- Russian Scientific and Research Institute of Hematology and Transfusiology, St Petersburg, Russia
| | | | - Munci Yagci
- Gazi Universitesi Tip Fakultesi, Ankara, Türkiye
| | - Loic Ysebaert
- Institut Universitaire du Cancer Toulouse Oncopole, Toulouse, France
| | - Keqin Qi
- Janssen Research & Development, Titusville, NJ, USA
| | - Qianya Qi
- Janssen Research & Development, Raritan, NJ, USA
| | - Pierre Sinet
- Janssen Research & Development, Bridgewater, NJ, USA
| | - Lori Parisi
- Janssen Research & Development, Raritan, NJ, USA
| | - Srimathi Srinivasan
- Oncology Translational Research, Janssen Research & Development, Lower Gwynedd Township, PA, USA
| | | | - Kurt Baeten
- Janssen Research & Development, Beerse, Belgium
| | - Angela Howes
- Janssen Research & Development, High Wycombe, UK
| | | | - Arnon P Kater
- Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
4
|
Fakhri B, Danilov A. SOHO State of the Art Updates and Next Questions: New Targetable Pathways in Chronic Lymphocytic Leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:232-237. [PMID: 36754692 DOI: 10.1016/j.clml.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
Regulatory approvals of Bruton tyrosine kinase (BTK) inhibitors and BCL2 inhibitors have transformed the therapeutic paradigm in chronic lymphocytic leukemia (CLL). However, despite significant improvement, treatment discontinuations due to an acquired resistance mutation or intolerance to these agents are common. Those who are refractory and/or intolerant to both these classes of drugs - the "double exposed/refractory" patients - pose a real challenge in clinical practice and are in dire need of novel therapeutic approaches. In this manuscript, we review the ongoing efforts addressing this unmet clinical need including the ongoing development of non-covalent BTK inhibitors, BTK degraders, novel BH3-mimetics, therapeutic antibodies targeting novel antigens and immune cell enabling therapies.
Collapse
Affiliation(s)
- Bita Fakhri
- Division of Hematology, Department of Medicine, Stanford University, Palo Alto, CA
| | - Alexey Danilov
- Department of Hematology and Hematopoietic Stem Cell Transplant, City of Hope National Medical Center, Duarte, CA.
| |
Collapse
|
5
|
Torka P, Thiruvengadam SK, Chen L, Wang X, Chen C, Vuong D, Qin H, Muir A, Orand K, Borja I, Lynne Smith D, Herrera AF, Spurgeon SEF, Park B, Lewis LD, Hernandez-Ilizaliturri F, Xia Z, Danilov AV. Pevonedistat, a Nedd8-activating enzyme inhibitor, in combination with ibrutinib in patients with relapsed/refractory B-cell non-Hodgkin lymphoma. Blood Cancer J 2023; 13:9. [PMID: 36631449 PMCID: PMC9834208 DOI: 10.1038/s41408-022-00763-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/27/2022] [Accepted: 11/29/2022] [Indexed: 01/12/2023] Open
Abstract
Pevonedistat (TAK924) is a Nedd8-activating enzyme inhibitor with preclinical activity in non-Hodgkin lymphoma (NHL). This open-label, Phase I, multicenter, investigator-sponsored study enrolled patients with relapsed/refractory (R/R) NHL and chronic lymphocytic leukemia (CLL). The primary objective was safety. Pevonedistat was given intravenously on days 1, 3, 5 of a 21-day cycle for 8 cycles at five dose levels (15 to 50 mg/m2); ibrutinib was administered at 420 or 560 mg orally daily continuously. Eighteen patients with NHL were enrolled, including 8 patients with mantle cell lymphoma (MCL) and 4 patients with CLL. One dose-limiting toxicity (mediastinal hemorrhage) occurred at 50 mg/m2 of pevonedistat which is the estimated maximum tolerated dose. Bruising and diarrhea were the most common adverse events (56% and 44%). Atrial fibrillation occurred in 3 patients (17%). Grade ≥3 toxicities included arthralgia, atrial fibrillation, bone pain, diarrhea, hypertension, and mediastinal hemorrhage (one patient each). The overall response rate (ORR) was 65% (100% ORR in MCL). Pevonedistat disposition was not modified by ibrutinib. scRNA-Seq analysis showed that pevonedistat downregulated NFκB signaling in malignant B-cells in vivo. Thus, pevonedistat combined with ibrutinib demonstrated safety and promising early efficacy in NHL and CLL. NAE inhibition downregulated NFκB signaling in vivo.
Collapse
Affiliation(s)
- Pallawi Torka
- Division of Hematology & Medical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | - Lu Chen
- City of Hope National Medical Center, Duarte, CA, USA
| | | | - Canping Chen
- Oregon Health and Science University, Portland, OR, USA
| | - Dan Vuong
- City of Hope National Medical Center, Duarte, CA, USA
| | - Hanjun Qin
- City of Hope National Medical Center, Duarte, CA, USA
| | | | - Kirsten Orand
- City of Hope National Medical Center, Duarte, CA, USA
| | - Ivana Borja
- City of Hope National Medical Center, Duarte, CA, USA
| | - D Lynne Smith
- City of Hope National Medical Center, Duarte, CA, USA
| | | | | | - Byung Park
- Oregon Health and Science University, Portland, OR, USA
| | - Lionel D Lewis
- Section of Clinical Pharmacology, Dept. of Medicine, The Geisel School of Medicine at Dartmouth and the Dartmouth Cancer Center, Lebanon, NH, USA
| | | | - Zheng Xia
- Oregon Health and Science University, Portland, OR, USA
| | | |
Collapse
|
6
|
EXABS-125-CLL Novel Agents in Chronic Lymphocytic Leukemia (CLL). CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22 Suppl 2:S30-S31. [PMID: 36163935 DOI: 10.1016/s2152-2650(22)00651-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
7
|
Kozyra P, Krasowska D, Pitucha M. New Potential Agents for Malignant Melanoma Treatment-Most Recent Studies 2020-2022. Int J Mol Sci 2022; 23:6084. [PMID: 35682764 PMCID: PMC9180979 DOI: 10.3390/ijms23116084] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 02/05/2023] Open
Abstract
Malignant melanoma (MM) is the most lethal skin cancer. Despite a 4% reduction in mortality over the past few years, an increasing number of new diagnosed cases appear each year. Long-term therapy and the development of resistance to the drugs used drive the search for more and more new agents with anti-melanoma activity. This review focuses on the most recent synthesized anti-melanoma agents from 2020-2022. For selected agents, apart from the analysis of biological activity, the structure-activity relationship (SAR) is also discussed. To the best of our knowledge, the following literature review delivers the latest achievements in the field of new anti-melanoma agents.
Collapse
Affiliation(s)
- Paweł Kozyra
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Danuta Krasowska
- Department of Dermatology, Venerology and Pediatric Dermatology, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| |
Collapse
|
8
|
Evans MG, Shestakova A, Haghighi N, Zhao X, Nardi V, Pinter-Brown LC, Rezk SA. Rare case of leptomeningeal small lymphocytic lymphoma with TP53 mutation detected by deep next-generation sequencing. Leuk Lymphoma 2022; 63:2479-2483. [DOI: 10.1080/10428194.2022.2070911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Mark G. Evans
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anna Shestakova
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Nahideh Haghighi
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Xiaohui Zhao
- Department of Pathology and Laboratory Medicine, University of California Irvine (UCI) Medical Center, Orange, CA, USA
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lauren C. Pinter-Brown
- Department of Medicine, Hematology/Oncology Division, University of California Irvine (UCI) Medical Center, Orange, CA, USA
| | - Sherif A. Rezk
- Department of Pathology and Laboratory Medicine, University of California Irvine (UCI) Medical Center, Orange, CA, USA
| |
Collapse
|
9
|
Dual BTK/SYK inhibition with CG-806 (luxeptinib) disrupts B-cell receptor and Bcl-2 signaling networks in mantle cell lymphoma. Cell Death Dis 2022; 13:246. [PMID: 35296646 PMCID: PMC8927405 DOI: 10.1038/s41419-022-04684-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 01/27/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022]
Abstract
Aberrant B-cell receptor (BCR) signaling is a key driver in lymphoid malignancies. Bruton tyrosine kinase (BTK) inhibitors that disrupt BCR signaling have received regulatory approvals in therapy of mantle cell lymphoma (MCL). However, responses are incomplete and patients who experience BTK inhibitor therapy failure have dire outcomes. CG-806 (luxeptinib) is a dual BTK/SYK inhibitor in clinical development in hematologic malignancies. Here we investigated the pre-clinical activity of CG-806 in MCL. In vitro treatment with CG-806 thwarted survival of MCL cell lines and patient-derived MCL cells in a dose-dependent manner. CG-806 blocked BTK and SYK activation and abrogated BCR signaling. Contrary to ibrutinib, CG-806 downmodulated the anti-apoptotic proteins Mcl-1 and Bcl-xL, abrogated survival of ibrutinib-resistant MCL cell lines, and partially reversed the pro-survival effects of stromal microenvironment-mimicking conditions in primary MCL cells. Dual BTK/SYK inhibition led to mitochondrial membrane depolarization accompanied by mitophagy and metabolic reprogramming toward glycolysis. In vivo studies of CG-806 demonstrated improved survival in one of the two tested aggressive MCL PDX models. While suppression of the anti-apoptotic Bcl-2 family proteins and NFκB signaling correlated with in vivo drug sensitivity, OxPhos and MYC transcriptional programs were upregulated in the resistant model following treatment with CG-806. BAX and NFKBIA were implicated in susceptibility to CG-806 in a whole-genome CRISPR-Cas9 library screen (in a diffuse large B-cell lymphoma cell line). A high-throughput in vitro functional drug screen demonstrated synergy between CG-806 and Bcl-2 inhibitors. In sum, dual BTK/SYK inhibitor CG-806 disrupts BCR signaling and induces metabolic reprogramming and apoptosis in MCL. The Bcl-2 network is a key mediator of sensitivity to CG-806 and combined targeting of Bcl-2 demonstrates synergy with CG-806 warranting continued exploration in lymphoid malignancies.
Collapse
|
10
|
Robak T, Witkowska M, Smolewski P. The Role of Bruton's Kinase Inhibitors in Chronic Lymphocytic Leukemia: Current Status and Future Directions. Cancers (Basel) 2022; 14:771. [PMID: 35159041 PMCID: PMC8833747 DOI: 10.3390/cancers14030771] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/24/2022] [Accepted: 01/31/2022] [Indexed: 12/20/2022] Open
Abstract
The use of Bruton's tyrosine kinase (BTK) inhibitors has changed the management and clinical history of patients with chronic lymphocytic leukemia (CLL). BTK is a critical molecule that interconnects B-cell antigen receptor (BCR) signaling. BTKis are classified into two categories: irreversible (covalent) inhibitors and reversible (non-covalent) inhibitors. Ibrutinib was the first irreversible BTK inhibitor approved by the U.S. Food and Drug Administration in 2013 as a breakthrough therapy in CLL patients. Subsequently, several studies have evaluated the efficacy and safety of new agents with reduced toxicity when compared with ibrutinib. Two other irreversible, second-generation BTK inhibitors, acalabrutinib and zanubrutinib, were developed to reduce ibrutinib-mediated adverse effects. Additionally, new reversible BTK inhibitors are currently under development in early-phase studies to improve their activity and to diminish adverse effects. This review summarizes the pharmacology, clinical efficacy, safety, dosing, and drug-drug interactions associated with the treatment of CLL with BTK inhibitors and examines their further implications.
Collapse
Affiliation(s)
- Tadeusz Robak
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland
| | - Magda Witkowska
- Department of Experimental Hematology, Medical University of Lodz, 93-510 Lodz, Poland; (M.W.); (P.S.)
| | - Piotr Smolewski
- Department of Experimental Hematology, Medical University of Lodz, 93-510 Lodz, Poland; (M.W.); (P.S.)
| |
Collapse
|
11
|
Xie H, Liu J, Alem Glison DM, Fleming JB. The clinical advances of proteolysis targeting chimeras in oncology. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:511-521. [PMID: 36046114 PMCID: PMC9400722 DOI: 10.37349/etat.2021.00061] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/20/2021] [Indexed: 12/18/2022] Open
Abstract
Proteolysis targeting chimeras (PROTACs) are a class of small molecules designed to target proteins for degradation. Their novel and unique modes of action provide PROTACs with the potential for their application in the management of both solid and hematologic malignancies. Since its initial discovery, the technology of targeted protein degradation, especially in the form of PROTACs, has had significant advances. A number of PROTACs have entered a late stage of preclinical development. Several of them are either in phase 1/2 clinical trials or approaching approval for initial clinical evaluation. This article discusses the preclinical and clinical findings of PROTACs of clinically relevant protein targets in cancer.
Collapse
Affiliation(s)
- Hao Xie
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Junjia Liu
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Diego M. Alem Glison
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jason B. Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
12
|
Ren S, Wang X, Song J, Jin G. Discovery of novel ibrutinib analogues to treat malignant melanoma. Bioorg Chem 2021; 117:105419. [PMID: 34689082 DOI: 10.1016/j.bioorg.2021.105419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/28/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022]
Abstract
A series of novel ibrutinib analogues was synthesized, and their proliferation inhibitory activities against various B lymphoma cell lines (DaudiB and Raji) and solid tumor cells (B16, CT26, HepG2 and 4T1) were evaluated. The most potent compound, YL7, exhibited strong antiproliferative activity in all cell lines, and its IC50 value in B16 cells was almost 9-fold better than that of ibrutinib. Mechanism of action studies showed that YL7 inhibited proliferation and migration and induced G1 cell cycle arrest, apoptosis and autophagy in B16 cells. Further assessment of in vivo antitumor efficacies demonstrated that YL7 significantly inhibited the growth of B16 melanoma. These preliminary studies suggest that it is reasonable to modify the structure of ibrutinib for antimelanoma treatment.
Collapse
Affiliation(s)
- Sumei Ren
- School of Pharmaceutical Sciences, Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, Shenzhen University Health Science Center, Shenzhen University, Shenzhen 518060, Guangdong, China; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xiaodong Wang
- School of Pharmaceutical Sciences, Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, Shenzhen University Health Science Center, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Jun Song
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Guangyi Jin
- School of Pharmaceutical Sciences, Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, Shenzhen University Health Science Center, Shenzhen University, Shenzhen 518060, Guangdong, China.
| |
Collapse
|
13
|
Multiparametric Flow Cytometry for MRD Monitoring in Hematologic Malignancies: Clinical Applications and New Challenges. Cancers (Basel) 2021; 13:cancers13184582. [PMID: 34572809 PMCID: PMC8470441 DOI: 10.3390/cancers13184582] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/05/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary In hematologic cancers, Minimal Residual Disease (MRD) monitoring, using either molecular (PCR) or immunophenotypic (MFC) diagnostics, allows the identification of rare cancer cells, readily detectable either in the bone marrow or in the peripheral blood at very low levels, far below the limit of classic microscopy. In this paper, we outlined the state-of-the-art of MFC-based MRD detection in different hematologic settings, highlighting main recommendations and new challenges for using such method in patients with acute leukemias or chronic hematologic neoplasms. The combination of new molecular technologies with advanced flow cytometry is progressively allowing clinicians to design a personalized therapeutic path, proportionate to the biological aggressiveness of the disease, in particular by using novel immunotherapies, in view of a modern decision-making process, based on precision medicine. Abstract Along with the evolution of immunophenotypic and molecular diagnostics, the assessment of Minimal Residual Disease (MRD) has progressively become a keystone in the clinical management of hematologic malignancies, enabling valuable post-therapy risk stratifications and guiding risk-adapted therapeutic approaches. However, specific prognostic values of MRD in different hematological settings, as well as its appropriate clinical uses (basically, when to measure it and how to deal with different MRD levels), still need further investigations, aiming to improve standardization and harmonization of MRD monitoring protocols and MRD-driven therapeutic strategies. Currently, MRD measurement in hematological neoplasms with bone marrow involvement is based on advanced highly sensitive methods, able to detect either specific genetic abnormalities (by PCR-based techniques and next-generation sequencing) or tumor-associated immunophenotypic profiles (by multiparametric flow cytometry, MFC). In this review, we focus on the growing clinical role for MFC-MRD diagnostics in hematological malignancies—from acute myeloid and lymphoblastic leukemias (AML, B-ALL and T-ALL) to chronic lymphocytic leukemia (CLL) and multiple myeloma (MM)—providing a comparative overview on technical aspects, clinical implications, advantages and pitfalls of MFC-MRD monitoring in different clinical settings.
Collapse
|