1
|
Teoh PJ, Koh MY, Mitsiades C, Gooding S, Chng WJ. Resistance to immunomodulatory drugs in multiple myeloma: the cereblon pathway and beyond. Haematologica 2025; 110:1074-1091. [PMID: 39569422 PMCID: PMC12050938 DOI: 10.3324/haematol.2024.285636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024] Open
Abstract
Acquired resistance to immunomodulatory drugs (IMiD) remains a significant unmet need in the treatment landscape of multiple myeloma (MM). The cereblon (CRBN) pathway-dependent mechanisms are known to be vital contributors to IMiD resistance; however, they may account for only a small proportion. Recent research has unveiled additional mechanisms of acquired IMiD resistance that are independent of the CRBN pathway. In this review, we provide a comprehensive overview of the existing work on IMiD resistance in MM, focusing specifically on the emerging evidence of CRBN pathway-independent mechanisms. Finally, we discuss the plausible actionable strategies and outlook for IMiD-based therapies moving forward.
Collapse
Affiliation(s)
- Phaik Ju Teoh
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore; Division of Molecular and Cellular Oncology (MCO), Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, 02142.
| | - Mun Yee Koh
- Cancer Science Institute of Singapore, National University of Singapore, 117599
| | - Constantine Mitsiades
- Division of Molecular and Cellular Oncology (MCO), Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, 02142
| | - Sarah Gooding
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK; Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, OX1 2JD, UK; MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS
| | - Wee Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore; Division of Haematology, Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Health System, 119228.
| |
Collapse
|
2
|
Chen P, Lin H, Xue Y, Thomas M, Wang A, Li Y, Cheng Y. A population pharmacokinetic analysis to evaluate the impact of renal impairment on the pharmacokinetics of iberdomide. J Pharm Sci 2025; 114:1315-1325. [PMID: 39870177 DOI: 10.1016/j.xphs.2025.01.015] [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: 11/01/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Iberdomide, a novel potent cereblon E3 ligase modulator, is under investigation for multiple myeloma. This study assessed how renal impairment (RI) affects iberdomide pharmacokinetics (PK). Twenty-six subjects with varying renal function, including those with severe renal impairment and those requiring intermittent hemodialysis (IHD), received a single oral 1 mg dose of iberdomide. Plasma, urine, and dialysate samples were analyzed to evaluate the PK of iberdomide and its major active metabolite, M12. Data were subsequently pooled with PK data from four other clinical trials involving 354 patients to develop a parent - metabolite population PK model using nonlinear mixed-effects modeling to assess the impact of various degrees of RI on drug exposure. The population PK model effectively described the PK of iberdomide and M12, showing that normal, mild, and moderate RI had no significant impact on iberdomide PK exposure, whereas severe RI reduced total clearance and increased PK exposure of iberdomide and M12. Subjects with kidney failure on IHD had comparable total clearance and PK exposure to those with normal renal function. These findings systematically examined the effects of various degrees of RI on iberdomide PK and provide a basis for informing iberdomide dosing in patients with varying degrees of renal impairment.
Collapse
Affiliation(s)
- Ping Chen
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Hongxia Lin
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yongjun Xue
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Mark Thomas
- Early Clinical Development ICN, Bristol Myers Squibb, Princeton, NJ, USA
| | - Alice Wang
- Early Clinical Development ICN, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yan Li
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yiming Cheng
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, USA.
| |
Collapse
|
3
|
Eckmann J, Fauti T, Biehl M, Zabaleta A, Blanco L, Lelios I, Gottwald S, Rae R, Lechner S, Bayer C, Dekempe Q, Osl F, Carrié N, Kassem S, Lorenz S, Christopeit T, Carpy A, Bujotzek A, Bröske AM, Dekhtiarenko I, Attig J, Kunz L, Cremasco F, Adelfio R, Fertig G, Dengl S, Gassner C, Bormann F, Kirstenpfad C, Kraft T, Diggelmann S, Knobloch M, Hage C, Feddersen R, Heidkamp G, Pöschinger T, Mayoux M, Bernasconi L, Prosper F, Dumontet C, Martinet L, Leclair S, Xu W, Paiva B, Klein C, Umaña P. Forimtamig, a novel GPRC5D-targeting T-cell bispecific antibody with a 2+1 format, for the treatment of multiple myeloma. Blood 2025; 145:202-219. [PMID: 39476124 PMCID: PMC11738037 DOI: 10.1182/blood.2024025987] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/15/2024] [Indexed: 01/11/2025] Open
Abstract
ABSTRACT Despite several approved therapies, multiple myeloma (MM) remains an incurable disease with high unmet medical need. "Off-the-shelf" T-cell bispecific antibodies (TCBs) targeting B-cell maturation antigen (BCMA) and G protein-coupled receptor class C group 5 member D (GPRC5D) have demonstrated high objective response rates in heavily pretreated patients with MM; however, primary resistance, short duration of response, and relapse driven by antigen shift frequently occur. Although GPRC5D represents the most selective target in MM, recent findings indicate antigen loss occurs more frequently than with BCMA. Thus, anti-GPRC5D immunotherapies must hit hard during a short period of time. Here, we characterize forimtamig, a novel GPRC5D-targeting TCB with 2+1 format. Bivalent binding of forimtamig to GPRC5D confers higher affinity than classical 1+1 TCB formats correlating with formation of more stable immunological synapses and higher potency in tumor cell killing and T-cell activation. Using an orthotopic mouse model of MM, forimtamig recruited T effector cells to the bone marrow and induced rapid tumor killing even after the introduction of step-up dosing to mitigate cytokine release. Combination of forimtamig with standard-of-care agents including anti-CD38 antibodies, immunomodulatory drugs, and proteasome inhibitors improved depth and duration of response. The combination of forimtamig with novel therapeutic agents including BCMA TCB and cereblon E3 ligase modulatory drugs was potent and prevented occurrence of GPRC5D -negative tumor relapse. Forimtamig is currently being evaluated in phase 1 clinical trials in patients with relapsed and refractory MM for monotherapy and in combination treatments. This trial was registered at www.ClinicalTrials.gov as #NCT04557150.
Collapse
Affiliation(s)
- Jan Eckmann
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Tanja Fauti
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Marlene Biehl
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Aintzane Zabaleta
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | - Laura Blanco
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | - Iva Lelios
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Stefan Gottwald
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Richard Rae
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Stefanie Lechner
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Christa Bayer
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Quincy Dekempe
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Franz Osl
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Nadege Carrié
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Sahar Kassem
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Stefan Lorenz
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Tony Christopeit
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Alejandro Carpy
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Alexander Bujotzek
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Ann-Marie Bröske
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Iryna Dekhtiarenko
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Jan Attig
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Leo Kunz
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Floriana Cremasco
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Roberto Adelfio
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Georg Fertig
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Stefan Dengl
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Christian Gassner
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Felix Bormann
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Claudia Kirstenpfad
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Thomas Kraft
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Sarah Diggelmann
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Melanie Knobloch
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Carina Hage
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Romi Feddersen
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Gordon Heidkamp
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Thomas Pöschinger
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Maud Mayoux
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Felipe Prosper
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | - Charles Dumontet
- Cancer Research Center of Lyon, INSERM 1052/ Centre National de la Recherche Scientifique 5286/University of Lyon/Hospices Civils de Lyon, Lyon, France
| | - Ludovic Martinet
- Cancer Research Center of Toulouse, INSERM, Centre National de la Recherche Scientifique, Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Stéphane Leclair
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Wei Xu
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Munich, Germany
| | - Bruno Paiva
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | - Christian Klein
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Pablo Umaña
- Discovery Oncology, Roche Pharma Research and Early Development, Roche Innovation Center, Zurich, Switzerland
| |
Collapse
|
4
|
Moukalled N, Abou Dalle I, El Cheikh J, Ye Y, Malarad F, Mohty M, Bazarbachi A. The emerging role of melflufen and peptide-conjugates in multiple myeloma. Curr Opin Oncol 2024; 36:583-592. [PMID: 39246181 DOI: 10.1097/cco.0000000000001090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
PURPOSE OF REVIEW The past two decades have witnessed an impressive expansion in the treatment landscape of multiple myeloma, leading to significant improvements in progression-free; as well as overall survival. However, almost all patients still experience multiple relapses during their disease course, with biological and cytogenetic heterogeneity affecting response to subsequent treatments. The purpose of this review is to provide a historical background regarding the role of alkylating agents and an updated data regarding the use of peptide-drug conjugates such as melflufen for patients with multiple myeloma. RECENT FINDINGS The combination of daratumumab-melflufen-dexamethasone evaluated in the LIGHTHOUSE study showed a statistically significant improvement in progression-free survival compared to single-agent daratumumab (not reached vs. 4.9 months respectively; P = 0.0032), with improvement in overall response rate to 59% vs. 30% respectively; P = 0.03. SUMMARY There have been an interest in developing and utilizing peptide-drug conjugates such as melflufen for treatment of patients with multiple myeloma, especially in the relapsed setting given historical results with alkylating agents, the use of which has been limited by dose-related toxicities in a disease that remains largely incurable. Single agent melflufen initially showed promising results especially in specific subgroups of heavily pretreated patients before the decision to suspend all clinical trials evaluating this agent after results from the OCEAN phase 3 trial. Subsequent reported analyses especially for melflufen-based combinations appear promising and suggest a potential use of peptide-drug conjugates provided optimal patient selection, as well as identification of the best companion agent.
Collapse
Affiliation(s)
- Nour Moukalled
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Iman Abou Dalle
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Jean El Cheikh
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yishan Ye
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Florent Malarad
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France
| | - Mohamad Mohty
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
5
|
Zheng X, Ji N, Campbell V, Slavin A, Zhu X, Chen D, Rong H, Enerson B, Mayo M, Sharma K, Browne CM, Klaus CR, Li H, Massa G, McDonald AA, Shi Y, Sintchak M, Skouras S, Walther DM, Yuan K, Zhang Y, Kelleher J, Liu G, Luo X, Mainolfi N, Weiss MM. Discovery of KT-474─a Potent, Selective, and Orally Bioavailable IRAK4 Degrader for the Treatment of Autoimmune Diseases. J Med Chem 2024; 67:18022-18037. [PMID: 39151120 PMCID: PMC11513890 DOI: 10.1021/acs.jmedchem.4c01305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/18/2024]
Abstract
Interleukin-1 receptor associated kinase 4 (IRAK4) is an essential mediator of the IL-1R and TLR signaling pathways, both of which have been implicated in multiple autoimmune conditions. Hence, blocking the activity of IRAK4 represents an attractive approach for the treatment of autoimmune diseases. The activity of this serine/threonine kinase is dependent on its kinase and scaffolding activities; thus, degradation represents a potentially superior approach to inhibition. Herein, we detail the exploration of structure-activity relationships that ultimately led to the identification of KT-474, a potent, selective, and orally bioavailable heterobifunctional IRAK4 degrader. This represents the first heterobifunctional degrader evaluated in a nononcology indication and dosed to healthy human volunteers. This molecule successfully completed phase I studies in healthy adult volunteers and patients with atopic dermatitis or hidradenitis suppurativa. Phase II clinical trials in both of these indications have been initiated.
Collapse
Affiliation(s)
- Xiaozhang Zheng
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Nan Ji
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Veronica Campbell
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Anthony Slavin
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Xiao Zhu
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Dapeng Chen
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Haojing Rong
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Brad Enerson
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Michele Mayo
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Kirti Sharma
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Chris M. Browne
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Christine R. Klaus
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Haoran Li
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Ginny Massa
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Alice A. McDonald
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Yatao Shi
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Mike Sintchak
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Stephanie Skouras
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Dirk M. Walther
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Karen Yuan
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Yi Zhang
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Joe Kelleher
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Guang Liu
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Xinbo Luo
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Nello Mainolfi
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| | - Matthew M. Weiss
- Kymera
Therapeutics, 500 N. Beacon Street, Fourth Floor, Watertown, Massachusetts 02472, United States
| |
Collapse
|
6
|
Brodermann MH, Henderson EK, Sellar RS. The emerging role of targeted protein degradation to treat and study cancer. J Pathol 2024; 263:403-417. [PMID: 38886898 DOI: 10.1002/path.6301] [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: 01/18/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 06/20/2024]
Abstract
The evolution of cancer treatment has provided increasingly targeted strategies both in the upfront and relapsed disease settings. Small-molecule inhibitors and immunotherapy have risen to prominence with chimeric antigen receptor T-cells, checkpoint inhibitors, kinase inhibitors, and monoclonal antibody therapies being deployed across a range of solid organ and haematological malignancies. However, novel approaches are required to target transcription factors and oncogenic fusion proteins that are central to cancer biology and have generally eluded successful drug development. Thalidomide analogues causing protein degradation have been a cornerstone of treatment in multiple myeloma, but a lack of in-depth mechanistic understanding initially limited progress in the field. When the protein cereblon (CRBN) was found to mediate thalidomide analogues' action and CRBN's neo-targets were identified, existing and novel drug development accelerated, with applications outside multiple myeloma, including non-Hodgkin's lymphoma, myelodysplastic syndrome, and acute leukaemias. Critically, transcription factors were the first canonical targets described. In addition to broadening the application of protein-degrading drugs, resistance mechanisms are being overcome and targeted protein degradation is widening the scope of druggable proteins against which existing approaches have been ineffective. Examples of targeted protein degraders include molecular glues and proteolysis targeting chimeras (PROTACs): heterobifunctional molecules that bind to proteins of interest and cause proximity-induced ubiquitination and proteasomal degradation via a linked E3 ligase. Twenty years since their inception, PROTACs have begun progressing through clinical trials, with early success in targeting the oestrogen receptor and androgen receptor in breast and prostate cancer respectively. This review explores important developments in targeted protein degradation to both treat and study cancer. It also considers the potential advantages and challenges in the translational aspects of developing new treatments. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Elizabeth K Henderson
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| | - Rob S Sellar
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
7
|
Liu Y, Mo CC, Hartley-Brown MA, Sperling AS, Midha S, Yee AJ, Bianchi G, Piper C, Tattersall A, Nadeem O, Laubach JP, Richardson PG. Targeting Ikaros and Aiolos: reviewing novel protein degraders for the treatment of multiple myeloma, with a focus on iberdomide and mezigdomide. Expert Rev Hematol 2024; 17:445-465. [PMID: 39054911 DOI: 10.1080/17474086.2024.2382897] [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: 03/21/2024] [Revised: 05/30/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION The treatment of multiple myeloma (MM) is evolving rapidly. Quadruplet regimens incorporating proteasome inhibitors, immunomodulatory drugs (IMiDs), and CD38 monoclonal antibodies have emerged as standard-of-care options for newly diagnosed MM, and numerous novel therapies have been approved for relapsed/refractory MM. However, there remains a need for novel options in multiple settings, including refractoriness to frontline standards of care. AREAS COVERED Targeting degradation of IKZF1 and IKZF3 - Ikaros and Aiolos - through modulation of cereblon, an E3 ligase substrate recruiter/receptor, is a key mechanism of action of the IMiDs and the CELMoD agents. Two CELMoD agents, iberdomide and mezigdomide, have demonstrated substantial preclinical and clinical activity in MM and have entered phase 3 investigation. Using a literature search methodology comprising searches of PubMed (unlimited time-frame) and international hematology/oncology conference abstracts (2019-2023), this paper reviews the importance of Ikaros and Aiolos in MM, the mechanism of action of the IMiDs and CELMoD agents and their relative potency for targeting Ikaros and Aiolos, and preclinical and clinical data on iberdomide and mezigdomide. EXPERT OPINION Emerging data suggest that iberdomide and mezigdomide have promising activity, including in IMiD-resistant settings and, pending phase 3 findings, may provide additional treatment options for patients with MM.
Collapse
Affiliation(s)
- Yuxin Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Clifton C Mo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Monique A Hartley-Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA
| | - Adam S Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA
| | - Shonali Midha
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA
| | - Andrew J Yee
- Massachusetts General Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Giada Bianchi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA
| | - Catherine Piper
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Alice Tattersall
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Omar Nadeem
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Jacob P Laubach
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Silva-Carvalho AÉ, Filiú-Braga LDC, Bogéa GMR, de Assis AJB, Pittella-Silva F, Saldanha-Araujo F. GLP and G9a histone methyltransferases as potential therapeutic targets for lymphoid neoplasms. Cancer Cell Int 2024; 24:243. [PMID: 38997742 PMCID: PMC11249034 DOI: 10.1186/s12935-024-03441-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024] Open
Abstract
Histone methyltransferases (HMTs) are enzymes that regulate histone methylation and play an important role in controlling transcription by altering the chromatin structure. Aberrant activation of HMTs has been widely reported in certain types of neoplastic cells. Among them, G9a/EHMT2 and GLP/EHMT1 are crucial for H3K9 methylation, and their dysregulation has been associated with tumor initiation and progression in different types of cancer. More recently, it has been shown that G9a and GLP appear to play a critical role in several lymphoid hematologic malignancies. Importantly, the key roles played by both enzymes in various diseases made them attractive targets for drug development. In fact, in recent years, several groups have tried to develop small molecule inhibitors targeting their epigenetic activities as potential anticancer therapeutic tools. In this review, we discuss the physiological role of GLP and G9a, their oncogenic functions in hematologic malignancies of the lymphoid lineage, and the therapeutic potential of epigenetic drugs targeting G9a/GLP for cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Alan Jhones Barbosa de Assis
- Laboratory of Molecular Pathology of Cancer, Faculty of Health Sciences and Medicine, University of Brasilia, Brasília, Brazil
| | - Fábio Pittella-Silva
- Laboratory of Molecular Pathology of Cancer, Faculty of Health Sciences and Medicine, University of Brasilia, Brasília, Brazil
| | - Felipe Saldanha-Araujo
- Hematology and Stem Cells Laboratory, Faculty of Health Sciences, University of Brasília, Brasilia, Brazil.
| |
Collapse
|
9
|
Schütt J, Brinkert K, Plis A, Schenk T, Brioli A. Unraveling the complexity of drug resistance mechanisms to SINE, T cell-engaging therapies and CELMoDs in multiple myeloma: a comprehensive review. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:26. [PMID: 39050883 PMCID: PMC11267153 DOI: 10.20517/cdr.2024.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/30/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024]
Abstract
Despite significant advances in the understanding of multiple myeloma (MM) biology and the development of novel treatment strategies in the last two decades, MM is still an incurable disease. Novel drugs with alternative mechanisms of action, such as selective inhibitors of nuclear export (SINE), modulators of the ubiquitin pathway [cereblon E3 ligase modulatory drugs (CELMoDs)], and T cell redirecting (TCR) therapy, have led to significant improvement in patient outcomes. However, resistance still emerges, posing a major problem for the treatment of myeloma patients. This review summarizes current data on treatment with SINE, TCR therapy, and CELMoDs and explores their mechanism of resistance. Understanding these resistance mechanisms is critical for developing strategies to overcome treatment failure and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Jacqueline Schütt
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Authors contributed equally
| | - Kerstin Brinkert
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Authors contributed equally
| | - Andrzej Plis
- Clinic for Internal Medicine C, Hematology and Oncology, Greifswald University Medicine, Greifswald 17489, Germany
| | - Tino Schenk
- Clinic of Internal Medicine 2, Department of Hematology and Medical Oncology, Jena University Hospital, Jena 07741, Germany
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena 07741, Germany
| | - Annamaria Brioli
- Clinic for Hematology, Hemostasis, Oncology and Stem cell transplantation, Hannover Medical School, Hannover 30625, Germany
- Clinic for Internal Medicine C, Hematology and Oncology, Greifswald University Medicine, Greifswald 17489, Germany
| |
Collapse
|
10
|
Amatangelo M, Flynt E, Stong N, Ray P, Van Oekelen O, Wang M, Ortiz M, Maciag P, Peluso T, Parekh S, van de Donk NWCJ, Lonial S, Thakurta A. Pharmacodynamic changes in tumor and immune cells drive iberdomide's clinical mechanisms of activity in relapsed and refractory multiple myeloma. Cell Rep Med 2024; 5:101571. [PMID: 38776914 PMCID: PMC11228401 DOI: 10.1016/j.xcrm.2024.101571] [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: 08/23/2023] [Revised: 03/20/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Iberdomide is a next-generation cereblon (CRBN)-modulating agent in the clinical development in multiple myeloma (MM). The analysis of biomarker samples from relapsed/refractory patients enrolled in CC-220-MM-001 (ClinicalTrials.gov: NCT02773030), a phase 1/2 study, shows that iberdomide treatment induces significant target substrate degradation in tumors, including in immunomodulatory agent (IMiD)-refractory patients or those with low CRBN levels. Additionally, some patients with CRBN genetic dysregulation who responded to iberdomide have a similar median progression-free survival (PFS) (10.9 months) and duration of response (DOR) (9.5 months) to those without CRBN dysregulation (11.2 month PFS, 9.4 month DOR). Iberdomide treatment promotes a cyclical pattern of immune stimulation without causing exhaustion, inducing a functional shift in T cells toward an activated/effector memory phenotype, including in triple-class refractory patients and those receiving IMiDs as a last line of therapy. This analysis demonstrates that iberdomide's clinical mechanisms of action are driven by both its cell-autonomous effects overcoming CRBN dysregulation in MM cells, and potent immune stimulation that augments anti-tumor immunity.
Collapse
Affiliation(s)
| | - Erin Flynt
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, USA
| | - Nicholas Stong
- Predictive Sciences, Bristol Myers Squibb, Summit, NJ, USA
| | - Pradipta Ray
- Data Sciences, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Oliver Van Oekelen
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Wang
- Translational Research, Bristol Myers Squibb, San Diego, CA, USA
| | - Maria Ortiz
- Predictive Sciences, BMS Center for Innovation and Translational Research Europe (CITRE), A Bristol Myers Squibb Company, Sevilla, Spain
| | - Paulo Maciag
- Clinical Development, Bristol Myers Squibb, Summit, NJ, USA
| | - Teresa Peluso
- Clinical Development, Bristol Myers Squibb, Summit, NJ, USA
| | - Samir Parekh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Niels W C J van de Donk
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, the Netherlands
| | - Sagar Lonial
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Anjan Thakurta
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, USA; Oxford Translational Myeloma Centre (OTMC), Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
11
|
Lin CHT, Tariq MJ, Ullah F, Sannareddy A, Khalid F, Abbas H, Bader A, Samaras C, Valent J, Khouri J, Anwer F, Raza S, Dima D. Current Novel Targeted Therapeutic Strategies in Multiple Myeloma. Int J Mol Sci 2024; 25:6192. [PMID: 38892379 PMCID: PMC11172591 DOI: 10.3390/ijms25116192] [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: 04/27/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy caused by the clonal expansion of immunoglobulin-producing plasma cells in the bone marrow and/or extramedullary sites. Common manifestations of MM include anemia, renal dysfunction, infection, bone pain, hypercalcemia, and fatigue. Despite numerous recent advancements in the MM treatment paradigm, current therapies demonstrate limited long-term effectiveness and eventual disease relapse remains exceedingly common. Myeloma cells often develop drug resistance through clonal evolution and alterations of cellular signaling pathways. Therefore, continued research of new targets in MM is crucial to circumvent cumulative drug resistance, overcome treatment-limiting toxicities, and improve outcomes in this incurable disease. This article provides a comprehensive overview of the landscape of novel treatments and emerging therapies for MM grouped by molecular target. Molecular targets outlined include BCMA, GPRC5D, FcRH5, CD38, SLAMF7, BCL-2, kinesin spindle protein, protein disulfide isomerase 1, peptidylprolyl isomerase A, Sec61 translocon, and cyclin-dependent kinase 6. Immunomodulatory drugs, NK cell therapy, and proteolysis-targeting chimera are described as well.
Collapse
Affiliation(s)
- Cindy Hsin-Ti Lin
- Department of Internal Medicine, Case Western Reserve University, MetroHealth Campus, Cleveland, OH 44109, USA
| | - Muhammad Junaid Tariq
- Department of Hematology-Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA;
| | - Fauzia Ullah
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | | | - Farhan Khalid
- Department of Internal Medicine, Monmouth Medical Center, Long Branch, NJ 07740, USA;
| | - Hasan Abbas
- Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Abbas Bader
- School of Medicine, University of Missouri–Kansas City, Kansas City, MO 64110, USA;
| | - Christy Samaras
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Jason Valent
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Jack Khouri
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Faiz Anwer
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Shahzad Raza
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
| | - Danai Dima
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA; (F.U.); (C.S.); (J.V.); (J.K.); (F.A.); (S.R.); (D.D.)
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
12
|
Ayto R, Annibali O, Biedermann P, Roset M, Sánchez E, Kotb R. The EASEMENT study: A multicentre, observational, cross-sectional study to evaluate patient preferences, treatment satisfaction, quality of life, and healthcare resource use in patients with multiple myeloma receiving injectable-containing or fully oral therapies. Eur J Haematol 2024; 112:889-899. [PMID: 38389468 DOI: 10.1111/ejh.14180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 02/24/2024]
Abstract
OBJECTIVE As multiple myeloma (MM) therapies advance, understanding patients', caregivers', and physicians' perspectives on, and satisfaction with, available treatment options and their impact on quality of life (QoL), is important. METHODS EASEMENT is a real-world, observational, cross-sectional study conducted in 19 sites within the UK, Canada, and Italy using retrospective chart reviews and surveys. Enrolled patients had clinical history available since diagnosis and had received ≥1 cycle of their current line of therapy. Primary objectives were to describe patient/caregiver QoL (EQ-5D-5L questionnaire), patient preference for oral/injectable therapies (single discrete-choice question), and patient satisfaction (TSQM-9 questionnaire). RESULTS Between October 2018 and March 2020, 399 patients were enrolled (n = 192 newly diagnosed multiple myeloma [NDMM], n = 206 relapsed/refractory multiple myeloma [RRMM], n = 1 missing). Among NDMM and RRMM patients, 78%/22% and 42%/58% were receiving injectables/orals, respectively. Both NDMM and RRMM patients significantly preferred orals versus injectables (p < .0001). No significant differences were reported in treatment satisfaction or QoL, but treatment convenience favoured orals over injectables with near significance (p = .053). CONCLUSION MM patients perceived greater convenience and preference for orals versus injectables. Oral treatments are useful for patients who cannot or prefer not to travel to clinics, or cannot perform self-injection within the community.
Collapse
Affiliation(s)
- Robert Ayto
- Department of Haematology, Queen Alexandra Hospital, Portsmouth, UK
| | - Ombretta Annibali
- Hematology, Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio Medico di Roma, Rome, Italy
| | - Patricia Biedermann
- Medical Affairs, Europe & Canada, Takeda Pharmaceuticals International AG, Zurich, Switzerland
| | | | | | - Rami Kotb
- Medical Oncology & Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
13
|
Sadhupati DP, Lakshmisudha R, Chakravarthy KNK, Naidana PS. A standardized combination of Terminalia chebula and Withania somnifera extracts enhances immune function in adults: a pilot randomized, double-blind, placebo-controlled clinical study. Food Nutr Res 2024; 68:10297. [PMID: 38863743 PMCID: PMC11165258 DOI: 10.29219/fnr.v68.10297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/19/2024] [Accepted: 04/21/2024] [Indexed: 06/13/2024] Open
Abstract
Background The use of botanical medicine has been demonstrated as a potential strategy to manage or treat a variety of health issues. Terminalia chebula (Retz) fruit and Withania somnifera (L.) Dunal roots are important medicinal herbs described in Ayurveda and traditional therapy for diverse health benefits. Objective This pilot study aimed to evaluate the immune function-enhancing potential of a unique blend of T. chebula fruit and W. somnifera root extracts, LN20189, in healthy men and women. Methods Forty healthy volunteers (age: 35-60 years) were randomized into two groups receiving either LN20189 (500 mg per day) or a matched placebo over 28 consecutive days. The total T-cell population was the primary efficacy measure in this study. The secondary efficacy measures included counts of CD4, CD8, natural killer (NK) cells, serum levels of interleukin-2 (IL-2), interferon-gamma (IFN-γ), total immunoglobulin-G (IgG), and Immune Function Questionnaire (IFQ) scores. Safety parameter assessments were also conducted. Results Post-trial, in LN20189-supplemented subjects, T cells, CD4, NK cells count, and the CD4:CD8 ratio were increased by 9.32, 10.10, 19.91, and 17.43%, respectively, as compared to baseline. LN20189 supplementation increased serum IFN-γ and IgG levels by 14.57 and 27.09% from baseline and by 13.98 and 21.99%, compared to placebo, respectively. Also, the IFQ scores in the LN20189 group were 84.68% (vs. baseline) and 69.44% (vs. placebo) lower at the end of the trial. LN20189 improved the study volunteers' cellular and humoral immune functions. Conclusion In summary, LN20189 supplementation was found tolerable and improved the key cellular and humoral factors of the immune system and helped improve immune function of the trial volunteers.
Collapse
Affiliation(s)
| | | | | | - Partha Sarathy Naidana
- Department of Community Medicine, ASR Academy of Medical Sciences, Eluru, Andhra Pradesh, India
| |
Collapse
|
14
|
Garfall AL. New Biological Therapies for Multiple Myeloma. Annu Rev Med 2024; 75:13-29. [PMID: 37729027 DOI: 10.1146/annurev-med-050522-033815] [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] [Indexed: 09/22/2023]
Abstract
Multiple myeloma is a cancer of bone marrow plasma cells that represents approximately 10% of hematologic malignancies. Though it is typically incurable, a remarkable suite of new therapies developed over the last 25 years has enabled durable disease control in most patients. This article briefly introduces the clinical features of multiple myeloma and aspects of multiple myeloma biology that modern therapies exploit. Key current and emerging treatment modalities are then reviewed, including cereblon-modulating agents, proteasome inhibitors, monoclonal antibodies, other molecularly targeted therapies (selinexor, venetoclax), chimeric antigen receptor T cells, T cell-engaging bispecific antibodies, and antibody-drug conjugates. For each modality, mechanism of action and clinical considerations are discussed. These therapies are combined and sequenced in modern treatment pathways, discussed at the conclusion of the article, which have led to substantial improvements in outcomes for multiple myeloma patients in recent years.
Collapse
Affiliation(s)
- Alfred L Garfall
- Division of Hematology/Oncology, Department of Medicine and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
15
|
Prabhala R, Pierceall WE, Samur M, Potluri LB, Hong K, Peluso T, Talluri S, Wang A, Katiki A, Vangala SD, Buonopane M, Bade V, Seah H, Krogman A, Derebail S, Fulciniti M, Lazo SB, Richardson P, Anderson K, Corre J, Avet-Loiseau H, Thakurta A, Munshi N. Immunomodulation of NK, NKT and B/T cell subtypes in relapsed/refractory multiple myeloma patients treated with pomalidomide along with velcade and dexamethasone and its association with improved progression-free survival. Front Oncol 2023; 13:1271807. [PMID: 38111533 PMCID: PMC10726115 DOI: 10.3389/fonc.2023.1271807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/08/2023] [Indexed: 12/20/2023] Open
Abstract
Background Multiple Myeloma (MM) patients exhibit dysregulated immune system, which is further weakened by chemotherapeutic agents. While cereblon-modulating agents, such as pomalidomide and lenalidomide, have been found to improve the immune profile, the efficacy of their impact in combination with other treatments is yet unknown. Methods We conducted an immune-profiling of a longitudinal cohort of 366 peripheral blood samples from the CC4047-MM-007 (OPTIMISMM, NCT01734928) study. This study followed relapsed/refractory Multiple Myeloma (RRMM) patients who were treated with Velcade + dexamethasone (Vd), or Vd with pomalidomide (PVd). 366 blood samples from 186 patients were evaluated using multi-color flow cytometry at 3 timepoints: screening, day 8 of cycle 1, and cycle 3. Results Among NK and NKT cell populations, adding pomalidomide showed no inhibition in the frequency of NK cells. When expression of double positivity for activation markers like, p46/NKG2D, on NK cells was higher than the median, PVd treated patients showed significantly better (p=0.05) progression-free survival (PFS) (additional 15 months) than patients with lower than the median expression of p46/NKG2D on NK cells. PVd treated patients who expressed CD158a/b below the median at cycle 1 demonstrated a significantly better PFS (more than 18months). Among B cell subtypes, PVd treatment significantly increased the abundance of B1b cells (p<0.05) and decreased Bregs (p<0.05) at day 8 of both cycle 1 and cycle 3 when compared to screening samples. Of all the B cell-markers evaluated among paired samples, a higher expression of MZB cells at day 8 of cycle 1 has resulted in enhanced PFS in PVd treated patients. Within T cells, pomalidomide treatment did not decrease the frequency of CD8 T cells when compared with screening samples. The higher the surface expression of OX-40 on CD8 T cells and the lower the expression of PD-1 and CD25 on CD4 T cells by PVd treatment resulted in improved PFS. Conclusion The prognostic significance for the number of immune markers is only seen in the PVd arm and none of these immune markers exhibit prognostic values in the Vd arm. This study demonstrates the importance of the immunomodulatory effects and the therapeutic benefit of adding pomalidomide to Vd treatment.
Collapse
Affiliation(s)
- Rao Prabhala
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | | | - Mehmet Samur
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, United States
| | - Lakshmi B. Potluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Kevin Hong
- Bristol Myers Squibb, Summit, NJ, United States
| | | | - Srikanth Talluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Angela Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Aishwarya Katiki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Sahan D. Vangala
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Michael Buonopane
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Vaishnavi Bade
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Hannah Seah
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Arthur Krogman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Sanika Derebail
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Mariateresa Fulciniti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Suzan B. Lazo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Paul Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Kenneth Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Jill Corre
- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | | | - Anjan Thakurta
- Bristol Myers Squibb, Summit, NJ, United States
- Oxford Centre for Translational Myeloma Research, Oxford, United Kingdom
| | - Nikhil Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
16
|
Neri P, Nijhof I. Evidence-based mechanisms of synergy with IMiD agent-based combinations in multiple myeloma. Crit Rev Oncol Hematol 2023:104041. [PMID: 37268176 DOI: 10.1016/j.critrevonc.2023.104041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 06/04/2023] Open
Abstract
Treatment of multiple myeloma (MM) has seen great advances in recent years, and a key contributor to this change has been the effective use of combination therapies, which have improved both the depth and duration of patient responses. IMiD agents (lenalidomide and pomalidomide) have both tumoricidal and immunostimulatory functions, and due to their multiple mechanisms of action have become the backbone of numerous combination treatments in the newly diagnosed and relapsed/refractory settings. Although IMiD agent-based combination regimens provide improved clinical outcomes for patients with MM, the mechanisms underpinning these combinations are not well understood. In this review we describe the potential mechanisms of synergy leading to the enhanced activity observed when IMiD agents and other drug classes are used in combination through interrogation of the current knowledge surrounding their mechanism of actions.
Collapse
Affiliation(s)
- Paola Neri
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada.
| | - Inger Nijhof
- Department of Hematology, Amsterdam University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands; Department of Internal Medicine and Department of Hematology, St. Antonius Hospital Nieuwegein, Koekoekslaan 1, 3435CM, Nieuwegein, the Netherlands
| |
Collapse
|
17
|
Wu Y, Shi X, Yao X, Du X. Biological research on the occurrence and development of multiple myeloma and its treatment. Immun Inflamm Dis 2023; 11:e850. [PMID: 37249283 PMCID: PMC10165958 DOI: 10.1002/iid3.850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/30/2023] [Accepted: 04/16/2023] [Indexed: 05/31/2023] Open
Abstract
INTRODUCTION To review the pathogenesis and treatment of multiple myeloma (MM). MM is a hematological malignancy with abnormal plasma cell proliferation in bone marrow. Due to the emergence of drug resistance, MM is still an incurable malignancy, which requires further exploration of pathogenesis and effective therapeutic targets. METHODS In this paper, the method of literature review is adopted to obtain the information about MM. Based on the literature, comprehensive and systematic review is made. RESULTS MM is a complex pathophysiological process with great heterogeneity, mainly reflected in genomic instability and bone marrow microenvironment. At present, the treatment of MM has made great progress, proteasome inhibitors and immunomodulatory drugs are widely used in clinic. Allogeneic stem cell transplantation may be the only promising cure for MM, and its high transplant-related mortality limits its clinical application. CONCLUSIONS The future of MM treatment lies in the development of more targeted therapies, novel immunotherapies, and a better understanding of the disease's molecular and genetic basis.
Collapse
Affiliation(s)
- Yue Wu
- Department of OrthopaedicsBeijing Chao‐Yang HospitalBeijingChina
| | - Xiangjun Shi
- Department of OrthopaedicsBeijing Chao‐Yang HospitalBeijingChina
| | - Xinchen Yao
- Department of OrthopaedicsBeijing Chao‐Yang HospitalBeijingChina
| | - Xinru Du
- Department of OrthopaedicsBeijing Chao‐Yang HospitalBeijingChina
| |
Collapse
|
18
|
Heim C, Spring AK, Kirchgäßner S, Schwarzer D, Hartmann MD. Cereblon neo-substrate binding mimics the recognition of the cyclic imide degron. Biochem Biophys Res Commun 2023; 646:30-35. [PMID: 36701892 DOI: 10.1016/j.bbrc.2023.01.051] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 01/17/2023] [Indexed: 01/19/2023]
Abstract
In targeted protein degradation, immunomodulatory drugs (IMiDs) or cereblon (CRBN) E3 ligase modulatory drugs (CELMoDs) recruit neo-substrate proteins to the E3 ubiquitin ligase receptor CRBN for ubiquitination and subsequent proteasomal degradation. While the structural basis of this mechanism is generally understood, we have only recently described the recognition mode of the natural CRBN degron. In this communication, we reveal that the IMiD- or CELMoD-mediated binding of neo-substrates closely mimics the recognition of natural degrons. In crystal structures, we identify a conserved binding mode for natural degron peptides with an elaborate hydrogen bonding network involving the backbone of each of the six C-terminal degron residues, without the involvement of side chains. In a structural comparison, we show that neo-substrates recruited by IMiDs or CELMoDs emulate every single hydrogen bond of this network and thereby explain the origins of the largely sequence-independent recognition of neo-substrates. Our results imply that the V388I substitution in CRBN does not impair natural degron recognition and complete the structural basis for the rational design of CRBN effectors.
Collapse
Affiliation(s)
- Christopher Heim
- Max Planck Institute for Biology, Tübingen, Germany; Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany; NanoTemper Technologies GmbH, Munich, Germany
| | | | - Sören Kirchgäßner
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Dirk Schwarzer
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Marcus D Hartmann
- Max Planck Institute for Biology, Tübingen, Germany; Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
19
|
Bhatt P, Kloock C, Comenzo R. Relapsed/Refractory Multiple Myeloma: A Review of Available Therapies and Clinical Scenarios Encountered in Myeloma Relapse. Curr Oncol 2023; 30:2322-2347. [PMID: 36826140 PMCID: PMC9954856 DOI: 10.3390/curroncol30020179] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
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).
Collapse
Affiliation(s)
- Parva Bhatt
- Correspondence: (P.B.); (R.C.); Tel.: +1-617-636-6454
| | | | | |
Collapse
|
20
|
Franz J, Myrus E, Sanchez L, Richter J. No needles needed: All-oral therapy options for relapsed and refractory multiple myeloma. Blood Rev 2023; 57:100993. [PMID: 36137842 DOI: 10.1016/j.blre.2022.100993] [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: 05/12/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 01/28/2023]
Abstract
Immense progress has been made for the treatment of multiple myeloma over the past two decades, with patient outcomes improving dramatically as a result. Patient quality of life, however, is constantly challenged by complications of the disease, side effects of therapy and the overall burden receiving continuous treatment. Compared to parenteral agents, all-oral regimens can provide logistically favorable alternatives and are associated with improved quality of life. Here, we review the currently available and investigational oral therapies for relapsed and refractory multiple myeloma and provide a practical clinical reference tool. We explore the factors that dictate the selection of therapy, such as prior drug refractoriness, disease biology and patient-specific considerations. Regimens with their respective supporting clinical data are organized by the degree of prior treatment, from lenalidomide-sensitive to heavily pretreated patients. We explore common challenges such as renal insufficiency and cytopenias. Lastly, we review investigational oral agents.
Collapse
Affiliation(s)
- Joseph Franz
- UPMC Hillman Cancer, University of Pittsburgh, Pittsburgh, PA, United States.
| | - Elizabeth Myrus
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Larysa Sanchez
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Joshua Richter
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
21
|
Cheng Y, Ye Y, Gaudy A, Ghosh A, Xue Y, Wang A, Zhou S, Li Y. A Phase 1, Multicenter, Open-Label Study to Evaluate the Pharmacokinetics of Iberdomide in Subjects with Mild, Moderate, or Severe Hepatic Impairment Compared with Healthy Subjects. Clin Pharmacol 2023; 15:9-19. [PMID: 36880014 PMCID: PMC9985425 DOI: 10.2147/cpaa.s397826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/17/2023] [Indexed: 03/04/2023] Open
Abstract
Introduction Iberdomide, a novel cereblon modulator (CELMoD®), is currently under clinical investigation for hematology indications. To evaluate the influence of hepatic impairment on the pharmacokinetics (PK) of iberdomide and its major active metabolite M12, a phase 1, multicenter, open-label study was conducted in healthy subjects and subjects with mild, moderate, and severe hepatic impairment. Methods Forty subjects were enrolled in the study and divided into five groups based on hepatic function. 1 mg iberdomide was administered and plasma samples were collected to evaluate the pharmacokinetics of iberdomide and M12. Results After a single dose of iberdomide (1 mg), mean iberdomide Cmax (maximum observed concentration) and AUC (area under the concentration-time curve) exposure were generally comparable between hepatic impairment (HI) subjects (severe, moderate and mild) and their respective matched normal controls. Mean Cmax and AUC exposure of the metabolite M12 were generally comparable between mild HI and matched normal subjects. However, mean Cmax of the M12 was 30% and 65% lower and AUC was 57% and 63% lower in moderate and severe HI subjects as compared to their respective matched normal controls. However, given the relatively low M12 exposure as compared to its parent drug, the observed differences were not considered clinically meaningful. Conclusion In summary, 1 mg single oral dose of iberdomide was generally well-tolerated. HI (mild, moderate or severe) had no clinically relevant impact on iberdomide PK and therefore, no dose adjustment is warranted.
Collapse
Affiliation(s)
- Yiming Cheng
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Ying Ye
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Allison Gaudy
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Atalanta Ghosh
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yongjun Xue
- Nonclinical Research & Development, Bristol Myers Squibb, Princeton, NJ, USA
| | - Alice Wang
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Simon Zhou
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yan Li
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
22
|
Cheng Y, Xue Y, Chen L, Masin M, Maciag P, Peluso T, Zhou S, Li Y. Model-based analysis for the population pharmacokinetics of iberdomide and its major active metabolite in healthy subjects and patients with relapsed and refractory multiple myeloma. Br J Clin Pharmacol 2023; 89:316-329. [PMID: 35981078 DOI: 10.1111/bcp.15498] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/25/2022] [Accepted: 08/15/2022] [Indexed: 11/28/2022] Open
Abstract
AIMS A parent-metabolite population pharmacokinetic (popPK) model of iberdomide and its pharmacologically active metabolite (M12) was developed and the influence of demographic and disease-related covariates on popPK parameters was assessed based on data from 3 clinical studies of iberdomide (dose range, 0.1-6 mg) in healthy subjects (n = 81) and patients with relapsed and refractory multiple myeloma (n 245). METHODS Nonlinear mixed effects modelling was used to develop the popPK model based on data from 326 subjects across 3 clinical studies. RESULTS The pharmacokinetics (PK) of iberdomide were adequately described with a 2-compartment model with first-order absorption and elimination. A first-order conversion rate was used to link the 1-compartment linear elimination metabolite model with the parent model. Subject type (multiple myeloma patients vs. healthy subject) was a statistically significant covariate on apparent clearance and apparent volume of distribution for the central compartment, suggesting different PK between patients with multiple myeloma and healthy subjects. Aspartate aminotransferase and sex were statistically but not clinically relevant covariates on apparent clearance. Metabolite (M12) PK tracked the PK of iberdomide. The metabolite to parent ratio was consistent across doses and combinations. CONCLUSION The parent-metabolite population PK model adequately described the time course PK data of iberdomide and M12. Iberdomide and M12 PK exposure were not complicated by demographic factors (age [19-82 y], body weight [41-172 kg], body surface area [1.4-2.7 m2 ], body mass index [16.4-59.3 kg/m2 ]), combination (in combination with dexamethasone and daratumumab), mild hepatic, or mild and moderate renal impairments. The model can be used to guide the dosing strategy for special patient population and inform future iberdomide study design.
Collapse
Affiliation(s)
- Yiming Cheng
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yongjun Xue
- Non-Clinical Research & Development, Bristol Myers Squibb, Princeton, NJ, USA
| | - Lu Chen
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Mark Masin
- Global Drug Development, Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | - Paulo Maciag
- Global Drug Development, Bristol Myers Squibb, Princeton, NJ, USA
| | - Teresa Peluso
- Global Drug Development, Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | - Simon Zhou
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| | - Yan Li
- Clinical Pharmacology & Pharmacometrics, Bristol Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
23
|
Fuchs O. Targeting cereblon in hematologic malignancies. Blood Rev 2023; 57:100994. [PMID: 35933246 DOI: 10.1016/j.blre.2022.100994] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
The protein cereblon (CRBN) is a substrate receptor of the cullin 4-really interesting new gene (RING) E3 ubiquitin ligase complex CRL4CRBN. Targeting CRBN mediates selective protein ubiquitination and subsequent degradation via the proteasome. This review describes novel thalidomide analogs, immunomodulatory drugs, also known as CRBN E3 ubiquitin ligase modulators or molecular glues (avadomide, iberdomide, CC-885, CC-90009, BTX-1188, CC-92480, CC-99282, CFT7455, and CC-91633), and CRBN-based proteolysis targeting chimeras (PROTACs) with increased efficacy and potent activity for application in hematologic malignancies. Both types of CRBN-binding drugs, molecular glues, and PROTACs stimulate the interaction between CRBN and its neosubstrates, recruiting target disease-promoting proteins and the E3 ubiquitin ligase CRL4CRBN. Proteins that are traditionally difficult to target (transcription factors and oncoproteins) can be polyubiquitinated and degraded in this way. The competition of CRBN neosubstrates with endogenous CRBN-interacting proteins and the pharmacology and rational combination therapies of and mechanisms of resistance to CRL4CRBN modulators or CRBN-based PROTACs are described.
Collapse
Affiliation(s)
- Ota Fuchs
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 12800 Praha 2, Czech Republic.
| |
Collapse
|
24
|
Melaccio A, Reale A, Saltarella I, Desantis V, Lamanuzzi A, Cicco S, Frassanito MA, Vacca A, Ria R. Pathways of Angiogenic and Inflammatory Cytokines in Multiple Myeloma: Role in Plasma Cell Clonal Expansion and Drug Resistance. J Clin Med 2022; 11:jcm11216491. [PMID: 36362718 PMCID: PMC9658666 DOI: 10.3390/jcm11216491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy, and despite the introduction of innovative therapies, remains an incurable disease. Identifying early and minimally or non-invasive biomarkers for predicting clinical outcomes and therapeutic responses is an active field of investigation. Malignant plasma cells (PCs) reside in the bone marrow (BM) microenvironment (BMME) which comprises cells (e.g., tumour, immune, stromal cells), components of the extracellular matrix (ECM) and vesicular and non-vesicular (soluble) molecules, all factors that support PCs’ survival and proliferation. The interaction between PCs and BM stromal cells (BMSCs), a hallmark of MM progression, is based not only on intercellular interactions but also on autocrine and paracrine circuits mediated by soluble or vesicular components. In fact, PCs and BMSCs secrete various cytokines, including angiogenic cytokines, essential for the formation of specialized niches called “osteoblastic and vascular niches”, thus supporting neovascularization and bone disease, vital processes that modulate the pathophysiological PCs–BMME interactions, and ultimately promoting disease progression. Here, we aim to discuss the roles of cytokines and growth factors in pathogenetic pathways in MM and as prognostic and predictive biomarkers. We also discuss the potential of targeted drugs that simultaneously block PCs’ proliferation and survival, PCs–BMSCs interactions and BMSCs activity, which may represent the future goal of MM therapy.
Collapse
Affiliation(s)
- Assunta Melaccio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.M.); (R.R.); Tel.: +39-320-55-17-232 (A.M.)
| | - Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne 3004, Australia
| | - Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Vanessa Desantis
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Aurelia Lamanuzzi
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Sebastiano Cicco
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Maria Antonia Frassanito
- General Pathology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.M.); (R.R.); Tel.: +39-320-55-17-232 (A.M.)
| |
Collapse
|
25
|
Gooding S, Ansari-Pour N, Kazeroun M, Karagoz K, Polonskaia A, Salazar M, Fitzsimons E, Sirinukunwattana K, Chavda S, Ortiz Estevez M, Towfic F, Flynt E, Pierceall W, Royston D, Yong K, Ramasamy K, Vyas P, Thakurta A. Loss of COP9 signalosome genes at 2q37 is associated with IMiD resistance in multiple myeloma. Blood 2022; 140:1816-1821. [PMID: 35853156 PMCID: PMC10653034 DOI: 10.1182/blood.2022015909] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/30/2022] [Indexed: 11/20/2022] Open
Abstract
The acquisition of a multidrug refractory state is a major cause of mortality in myeloma. Myeloma drugs that target the cereblon (CRBN) protein include widely used immunomodulatory drugs (IMiDs), and newer CRBN E3 ligase modulator drugs (CELMoDs), in clinical trials. CRBN genetic disruption causes resistance and poor outcomes with IMiDs. Here, we investigate alternative genomic associations of IMiD resistance, using large whole-genome sequencing patient datasets (n = 522 cases) at newly diagnosed, lenalidomide (LEN)-refractory and lenalidomide-then-pomalidomide (LEN-then-POM)-refractory timepoints. Selecting gene targets reproducibly identified by published CRISPR/shRNA IMiD resistance screens, we found little evidence of genetic disruption by mutation associated with IMiD resistance. However, we identified a chromosome region, 2q37, containing COP9 signalosome members COPS7B and COPS8, copy loss of which significantly enriches between newly diagnosed (incidence 5.5%), LEN-refractory (10.0%), and LEN-then-POM-refractory states (16.4%), and may adversely affect outcomes when clonal fraction is high. In a separate dataset (50 patients) with sequential samples taken throughout treatment, we identified acquisition of 2q37 loss in 16% cases with IMiD exposure, but none in cases without IMiD exposure. The COP9 signalosome is essential for maintenance of the CUL4-DDB1-CRBN E3 ubiquitin ligase. This region may represent a novel marker of IMiD resistance with clinical utility.
Collapse
Affiliation(s)
- Sarah Gooding
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Haematology, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
| | - Naser Ansari-Pour
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Mohammad Kazeroun
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Kubra Karagoz
- Translational Medicine, Bristol Myers Squibb, Summit, NJ
| | - Ann Polonskaia
- Translational Medicine, Bristol Myers Squibb, Summit, NJ
| | - Mirian Salazar
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
| | - Evie Fitzsimons
- Department of Haematology, Cancer Institute, University College London, United Kingdom
| | | | - Selina Chavda
- Department of Haematology, Cancer Institute, University College London, United Kingdom
| | - Maria Ortiz Estevez
- Bristol Myers Squibb Center for Innovation and Translational Research Europe, Sevilla, Spain
| | | | - Erin Flynt
- Translational Medicine, Bristol Myers Squibb, Summit, NJ
| | | | - Daniel Royston
- Nuffield Department of Cellular and Laboratory Sciences, University of Oxford, Oxford, United Kingdom
| | - Kwee Yong
- Department of Haematology, Cancer Institute, University College London, United Kingdom
| | - Karthik Ramasamy
- Department of Haematology, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
| | - Paresh Vyas
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Haematology, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Anjan Thakurta
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
26
|
CRL4 CRBN E3 Ligase Complex as a Therapeutic Target in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14184492. [PMID: 36139651 PMCID: PMC9496858 DOI: 10.3390/cancers14184492] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Immunomodulatory drugs (IMiDs) are effective in the treatment of multiple myeloma (MM) and other hematological malignancies. Cereblon (CRBN), a target of IMiDs, forms the CRL4 E3 ubiquitin ligase complex (CRL4CRBN) with DDB1, CUL4A and RBX1. The insight into the molecular mechanism of IMiDs action has advanced dramatically since the identification of cereblon (CRBN) as their direct target. Targeting CRBN by IMiDs modifies CRL4CRBN substrate specificity towards non-physiological protein targets which are subsequently ubiquitinated and degraded by the proteasome. To date, IMiDs are the only known group of protein degraders used in clinical practice. This review provides the current state of knowledge about thalidomide and its derivatives’ mechanisms of action, and highlights the future perspectives for targeted protein degraders. 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 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.
Collapse
|
27
|
Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers (Basel) 2022; 14:cancers14174082. [PMID: 36077618 PMCID: PMC9454959 DOI: 10.3390/cancers14174082] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.
Collapse
Affiliation(s)
- Danai Dima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongxu Jiang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Divya Jyoti Singh
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Haikoo S. Shah
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
28
|
Cheng Y, Hong K, Chen N, Yu X, Peluso T, Zhou S, Li Y. Aiding early clinical drug development by elucidation of the relationship between tumor growth inhibition and survival in relapsed/refractory multiple myeloma patients. EJHAEM 2022; 3:815-827. [PMID: 36051011 PMCID: PMC9422038 DOI: 10.1002/jha2.494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Early prognosis of clinical efficacy is an urgent need for oncology drug development. Herein, we systemically examined the quantitative approach of tumor growth inhibition (TGI) and survival modeling in the space of relapsed and refractory multiple myeloma (MM), aiming to provide insights into clinical drug development. Longitudinal serum M-protein and progression-free survival (PFS) data from three phase III studies (N = 1367) across six treatment regimens and different patient populations were leveraged. The TGI model successfully described the longitudinal M-protein data in patients with MM. The tumor inhibition and growth parameters were found to vary as per each study, likely due to the patient population and treatment regimen difference. Based on a parametric time-to-event model for PFS, M-protein reduction at week 4 was identified as a significant prognostic factor for PFS across the three studies. Other factors, including Eastern Cooperative Oncology Group performance status, prior anti-myeloma therapeutics, and baseline serum ß2-microglobulin level, were correlated with PFS as well. In conclusion, patient disease characteristics (i.e., baseline tumor burden and treatment lines) were important determinants of tumor inhibition and PFS in MM patients. M-protein change at week 4 was an early prognostic biomarker for PFS.
Collapse
Affiliation(s)
- Yiming Cheng
- Clinical Pharmacology & PharmacometricsBristol Myers SquibbNew JerseyUSA
| | - Kevin Hong
- Global Drug DevelopmentBristol Myers SquibbNew JerseyUSA
| | - Nianhang Chen
- Clinical Pharmacology & PharmacometricsBristol Myers SquibbNew JerseyUSA
| | - Xin Yu
- Global Biometric SciencesBristol Myers SquibbNew JerseyUSA
| | - Teresa Peluso
- Global Drug Development Bristol Myers SquibbBoudrySwitzerland
| | - Simon Zhou
- Clinical Pharmacology & PharmacometricsBristol Myers SquibbNew JerseyUSA
| | - Yan Li
- Clinical Pharmacology & PharmacometricsBristol Myers SquibbNew JerseyUSA
| |
Collapse
|
29
|
Costa BA, Mouhieddine TH, Richter J. What's Old is New: The Past, Present and Future Role of Thalidomide in the Modern-Day Management of Multiple Myeloma. Target Oncol 2022; 17:383-405. [PMID: 35771402 DOI: 10.1007/s11523-022-00897-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2022] [Indexed: 10/17/2022]
Abstract
Immunomodulatory drugs (IMiDs) have become an integral part of therapy for both newly diagnosed and relapsed/refractory multiple myeloma (RRMM). IMiDs bind to cereblon, leading to the degradation of proteins involved in B-cell survival and proliferation. Thalidomide, a first-generation IMiD, has little to no myelosuppressive potential, negligible renal clearance, and long-proven anti-myeloma activity. However, thalidomide's adverse effects (e.g., somnolence, constipation, and peripheral neuropathy) and the advent of more potent therapeutic options has led to the drug being less frequently used in many countries, including the US and Canada. Newer-generation IMiDs, such as lenalidomide and pomalidomide, are utilized far more frequently. In numerous previous trials, salvage therapy with thalidomide (50-200 mg/day) plus corticosteroids (with or without selected cytotoxic or targeted agents) has been shown to be effective and well-tolerated in the RRMM setting. Hence, thalidomide-based regimens remain important alternatives for heavily pretreated patients, especially for those who have no access to novel therapies and/or are not eligible for their use (due to renal failure, high-grade myelosuppression, or significant comorbidities). Ongoing and future trials may provide further insights into the current role of thalidomide, especially by comparing thalidomide-containing regimens with protocols based on newer-generation IMiDs and by investigating thalidomide's association with novel therapies (e.g., antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor T cells).
Collapse
Affiliation(s)
- Bruno Almeida Costa
- Department of Medicine, Mount Sinai Morningside and West, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tarek H Mouhieddine
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1185, New York, NY, 10029, USA
| | - Joshua Richter
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1185, New York, NY, 10029, USA.
| |
Collapse
|
30
|
Offidani M, Corvatta L, Morè S, Manieri MV, Olivieri A. An update on novel multiple myeloma targets. Expert Rev Hematol 2022; 15:519-537. [PMID: 35640130 DOI: 10.1080/17474086.2022.2085088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Introduction: despite therapeutic progress, leading to a significant improvement of outcome, multiple myeloma (MM) remains a difficult to treat hematologic disease due to its biological heterogeneity and clinical complexity. Areas covered: Treatment of patients refractory and resistant to all classes of agents used in newly diagnosed MM, is becoming a relevant problem for every hematologist. New generation immunotherapies, such as conjugated mAb, bispecific mAbs and CAR-T cells, targeting novel molecules as BCMA, have showed relevant results in very advanced MM. In the same setting, small molecules, such as selinexor and melflufen, also proved to be effective. We are currently waiting for the results of under evaluation personalized therapy, directed against specific gene mutations or signaling pathways, responsible for disease progression. Expert Opinion: In the near future, many therapeutic strategies will become available for MM and the challenge will be to position each approach in order to cure, maintaining a good quality of life in these patients.
Collapse
Affiliation(s)
- Massimo Offidani
- Clinica di Ematologia Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona
| | | | - Sonia Morè
- Clinica di Ematologia Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona
| | | | - Attilio Olivieri
- Clinica di Ematologia Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona
| |
Collapse
|
31
|
Richardson PG, Mateos MV, Vangsted AJ, Ramasamy K, Abildgaard N, Ho PJ, Quach H, Bahlis NJ. The role of E3 ubiquitin ligase in multiple myeloma: potential for cereblon E3 ligase modulators in the treatment of relapsed/refractory disease. Expert Rev Proteomics 2022; 19:235-246. [PMID: 36342226 DOI: 10.1080/14789450.2022.2142564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Insights into the mechanisms of protein homeostasis and proteasomal degradation have led to new strategies of redirecting the ubiquitin-proteasome system (UPS) to reduce or eliminate proteins or survival factors key to malignant pathobiology, multiple myeloma (MM) in particular. These strategies have enabled researchers to target proteins that were previously considered difficult to modulate by pharmacological means. AREAS COVERED This review provides a brief overview of UPS biology, particularly the role of the CRL4CRBN E3 ubiquitin ligase complex, and summarizes current strategies for co-opting the UPS, including CELMoD compounds, SNIPERs, PROTACs, and degronimids. A detailed discussion is provided on lead CELMoD compounds iberdomide and mezigdomide, which are currently being evaluated in clinical trials in patients with MM. EXPERT OPINION Since a high proportion of patients develop drug resistance, it is vital to have novel therapeutic agents for treating relapsed patients with MM more effectively. It is encouraging that the expanding pathophysiological insight into cellular signaling pathways in MM increasingly translates into the development of novel therapeutic agents such as targeted protein degraders. This holds promise for improving outcomes in MM and beyond.
Collapse
Affiliation(s)
- Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - Karthik Ramasamy
- Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Niels Abildgaard
- Hematology Research Unit, Department of Hematology, Odense University Hospital, Odense, Denmark; and Department of Clinical Research.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - P Joy Ho
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.,Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | - Hang Quach
- Department of Haematology, St Vincent's Hospital, Melbourne, Australia
| | - Nizar J Bahlis
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
32
|
Karagoz K, Stokes M, Ortiz-Estévez M, Towfic F, Flynt E, Gooding S, Pierceall W, Thakurta A. Multiple Myeloma Patient Tumors With High Levels of Cereblon Exon-10 Deletion Splice Variant Upregulate Clinically Targetable Pro-Inflammatory Cytokine Pathways. Front Genet 2022; 13:831779. [PMID: 35222546 PMCID: PMC8864318 DOI: 10.3389/fgene.2022.831779] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/14/2022] [Indexed: 12/13/2022] Open
Abstract
Immunomodulatory drugs (IMiDs), including lenalidomide and pomalidomide, are used in the routine treatment for multiple myeloma (MM) patients. Cereblon (CRBN) is the direct molecular target of IMiDs. While CRBN is not an essential gene for MM cell proliferation, the frequency of CRBN genetic aberrations, including mutation, copy number loss, and exon-10 (which includes a portion of the IMiD-binding domain) splicing, have been reported to incrementally increase in later-line patients. CRBN exon-10 splicing has also been shown to be associated with decreased progression-free survival in both newly diagnosed and relapsed refractory MM patients. Although we did not find significant general splicing defects among patients with CRBN exon-10 splice variant when compared to those expressing the full-length transcript, we identified upregulated TNFA signaling via NFKB, inflammatory response, and IL-10 signaling pathways in patients with exon-10 splice variant across various data sets—all potentially promoting tumor growth via chronic growth signals. We examined master regulators that mediate transcriptional programs in CRBN exon-10 splice variant patients and identified BATF, EZH2, and IKZF1 as the key candidates across the four data sets. Upregulated downstream targets of BATF, EZH2, and IKZF1 are components of TNFA signaling via NFKB, IL2/STAT5 signaling pathways, and IFNG response pathways. Previously, BATF-mediated transcriptional regulation was associated with venetoclax sensitivity in MM. Interestingly, we found that an EZH2 sensitivity gene expression signature also correlated with high BATF or venetoclax sensitivity scores in these tumors. Together, these data provide a rationale for investigating EZH2 inhibitors or venetoclax in combination with the next generation CRBN-targeting agents, such as CELMoDs, for patients overexpressing the CRBN exon-10 splice variant.
Collapse
Affiliation(s)
- Kubra Karagoz
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, United States
| | - Matthew Stokes
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, United States
| | - María Ortiz-Estévez
- Bristol Myers Squibb Center for Innovation and Translational Research Europe, Sevilla, Spain
| | - Fadi Towfic
- Bristol Myers Squibb, San Diego, CA, United States
| | - Erin Flynt
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, United States
| | - Sarah Gooding
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Department of Haematology, Oxford University Hospitals NHS Trust, Oxford, United Kingdom.,NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom.,Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
| | - William Pierceall
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, United States
| | - Anjan Thakurta
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, United States.,Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
33
|
Lee HF, Lacbay CM, Boutin R, Matralis AN, Park J, Waller DD, Guan TL, Sebag M, Tsantrizos YS. Synthesis and Evaluation of Structurally Diverse C-2-Substituted Thienopyrimidine-Based Inhibitors of the Human Geranylgeranyl Pyrophosphate Synthase. J Med Chem 2022; 65:2471-2496. [PMID: 35077178 DOI: 10.1021/acs.jmedchem.1c01913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Novel analogues of C-2-substituted thienopyrimidine-based bisphosphonates (C2-ThP-BPs) are described that are potent inhibitors of the human geranylgeranyl pyrophosphate synthase (hGGPPS). Members of this class of compounds induce target-selective apoptosis of multiple myeloma (MM) cells and exhibit antimyeloma activity in vivo. A key structural element of these inhibitors is a linker moiety that connects their (((2-phenylthieno[2,3-d]pyrimidin-4-yl)amino)methylene)bisphosphonic acid core to various side chains. The structural diversity of this linker moiety, as well as the side chains attached to it, was investigated and found to significantly impact the toxicity of these compounds in MM cells. The most potent inhibitor identified was evaluated in mouse and rat for liver toxicity and systemic exposure, respectively, providing further optimism for the potential value of such compounds as human therapeutics.
Collapse
Affiliation(s)
- Hiu-Fung Lee
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Cyrus M Lacbay
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Rebecca Boutin
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Alexios N Matralis
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Jaeok Park
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Daniel D Waller
- Department of Medicine, McGill University, Montreal, Quebec H3A 1A1, Canada
- Division of Hematology, McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Tian Lai Guan
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Michael Sebag
- Department of Medicine, McGill University, Montreal, Quebec H3A 1A1, Canada
- Division of Hematology, McGill University Health Center, Montreal, Quebec H4A 3J1, Canada
| | - Youla S Tsantrizos
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
34
|
Zhou S, Wang R. Targeted therapy of multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:465-480. [PMID: 36045700 PMCID: PMC9400694 DOI: 10.37349/etat.2021.00057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/28/2021] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM) is a malignant proliferative disease of monoclonal plasma cells (PCs) and is characterized by uncontrolled proliferation of PCs and excessive production of specific types of immunoglobulins. Since PCs are terminally differentiated B cells, the World Health Organization (WHO) classifies MM as lymphoproliferative B-cell disease. The incidence of MM is 6-7 cases per 100,000 people in the world every year and the second most common cancer in the blood system. Due to the effects of drug resistance and malignant regeneration of MM cells in the microenvironment, all current treatment methods can prolong both overall and symptom-free survival rates of patients with MM but cannot cure MM. Both basic and clinical studies have proven that targeted therapy leads to a clear and significant prolongation of the survival of patients with MM, but when the disease recurs again, resistance to the previous treatment will occur. Therefore, the discovery of new targets and treatment methods plays a vital role in the treatment of MM. This article introduces and summarizes targeted MM therapy, potential new targets, and future precision medicine in MM.
Collapse
Affiliation(s)
- Shan Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Renxi Wang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| |
Collapse
|
35
|
Sgherza N, Curci P, Rizzi R, Musto P. Novel Approaches Outside the Setting of Immunotherapy for the Treatment of Multiple Myeloma: The Case of Melflufen, Venetoclax, and Selinexor. Front Oncol 2021; 11:716751. [PMID: 34660279 PMCID: PMC8514936 DOI: 10.3389/fonc.2021.716751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/18/2021] [Indexed: 11/13/2022] Open
Abstract
Although the survival rate of patients with multiple myeloma has significantly improved in the last years thanks to the introduction of various classes of new drugs, such as proteasome inhibitors, immunomodulatory agents, and monoclonal antibodies, the vast majority of these subjects relapse with a more aggressive disease due to the acquisition of further genetic alterations that may cause resistance to current salvage therapies. The treatment of these often "triple" (or even more) refractory patients remains challenging, and alternative approaches are required to overcome the onset of that resistance. Immunotherapies with novel monoclonal, drug-conjugated, or bi-specific antibodies, as well as the use of chimeric antigen receptor T cells, have been recently developed and are currently investigated. However, other non-immunologic therapeutic regimens based on melfluflen, venetoclax, or selinexor, three molecules with new mechanisms of action, have also shown promising results in the setting of relapsed/refractory myeloma. Here we report the most recent literature data regarding these three drugs, focusing on their efficacy and safety in multiple myeloma.
Collapse
Affiliation(s)
- Nicola Sgherza
- Unit of Hematology and Stem Cell Transplantation, Azienda Ospedaliero Universitaria Consorziale (AOUC) Policlinico, Bari, Italy
| | - Paola Curci
- Unit of Hematology and Stem Cell Transplantation, Azienda Ospedaliero Universitaria Consorziale (AOUC) Policlinico, Bari, Italy
| | - Rita Rizzi
- Unit of Hematology and Stem Cell Transplantation, Azienda Ospedaliero Universitaria Consorziale (AOUC) Policlinico, Bari, Italy
- Department of Emergency and Organ Transplantation, “Aldo Moro” University School of Medicine, Bari, Italy
| | - Pellegrino Musto
- Unit of Hematology and Stem Cell Transplantation, Azienda Ospedaliero Universitaria Consorziale (AOUC) Policlinico, Bari, Italy
- Department of Emergency and Organ Transplantation, “Aldo Moro” University School of Medicine, Bari, Italy
| |
Collapse
|