51
|
Byun JY, Koh YT, Jang SY, Witcher JW, Chan JR, Pustilnik A, Daniels MJ, Kim YH, Suh KH, Linnik MD, Lee YM. Target modulation and pharmacokinetics/pharmacodynamics translation of the BTK inhibitor poseltinib for model-informed phase II dose selection. Sci Rep 2021; 11:18671. [PMID: 34548595 PMCID: PMC8455565 DOI: 10.1038/s41598-021-98255-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 09/01/2021] [Indexed: 01/14/2023] Open
Abstract
The selective Bruton tyrosine kinase (BTK) inhibitor poseltinib has been shown to inhibit the BCR signal transduction pathway and cytokine production in B cells (Park et al.Arthritis Res. Ther.18, 91, 10.1186/s13075-016-0988-z, 2016). This study describes the translation of nonclinical research studies to a phase I clinical trial in healthy volunteers in which pharmacokinetics (PKs) and pharmacodynamics (PDs) were evaluated for dose determination. The BTK protein kinase inhibitory effects of poseltinib in human peripheral blood mononuclear cells (PBMCs) and in rats with collagen-induced arthritis (CIA) were evaluated. High-dimensional phosphorylation analysis was conducted on human immune cells such as B cells, CD8 + memory cells, CD4 + memory cells, NK cells, neutrophils, and monocytes, to map the impact of poseltinib on BTK/PLC and AKT signaling pathways. PK and PD profiles were evaluated in a first-in-human study in healthy donors, and a PK/PD model was established based on BTK occupancy. Poseltinib bound to the BTK protein and modulated BTK phosphorylation in human PBMCs. High-dimensional phosphorylation analysis of 94 nodes showed that poseltinib had the highest impact on anti-IgM + CD40L stimulated B cells, however, lower impacts on anti-CD3/CD-28 stimulated T cells, IL-2 stimulated CD4 + T cells and NK cells, M-CSF stimulated monocytes, or LPS-induced granulocytes. In anti-IgM + CD40L stimulated B cells, poseltinib inhibited the phosphorylation of BTK, AKT, and PLCγ2. Moreover, poseltinib dose dependently improved arthritis disease severity in CIA rat model. In a clinical phase I trial for healthy volunteers, poseltinib exhibited dose-dependent and persistent BTK occupancy in PBMCs of all poseltinib-administrated patients in the study. More than 80% of BTK occupancy at 40 mg dosing was maintained for up to 48 h after the first dose. A first-in-human healthy volunteer study of poseltinib established target engagement with circulating BTK protein. Desirable PK and PD properties were observed, and a modeling approach was used for rational dose selection for subsequent trials. Poseltinib was confirmed as a potential BTK inhibitor for the treatment of autoimmune diseases. Trial registration: This article includes the results of a clinical intervention on human participants [NCT01765478].
Collapse
Affiliation(s)
- Joo-Yun Byun
- Hanmi Research Center, Hanmi Pharm. Co. Ltd., 14 Wiryeseong-daero, Songpa-gu, Seoul, 05545, Korea
| | - Yi T Koh
- Lilly Biotechnology Center, 10290 Campus Point Drive, San Diego, 92121, USA
| | - Sun Young Jang
- Hanmi Research Center, Hanmi Pharm. Co. Ltd., 14 Wiryeseong-daero, Songpa-gu, Seoul, 05545, Korea
| | - Jennifer W Witcher
- Lilly Biotechnology Center, 10290 Campus Point Drive, San Diego, 92121, USA
| | - Jason R Chan
- Lilly Biotechnology Center, 10290 Campus Point Drive, San Diego, 92121, USA
| | - Anna Pustilnik
- Lilly Biotechnology Center, 10290 Campus Point Drive, San Diego, 92121, USA
| | - Mark J Daniels
- Lilly Biotechnology Center, 10290 Campus Point Drive, San Diego, 92121, USA
| | - Young Hoon Kim
- Hanmi Research Center, Hanmi Pharm. Co. Ltd., 14 Wiryeseong-daero, Songpa-gu, Seoul, 05545, Korea
| | - Kwee Hyun Suh
- Hanmi Research Center, Hanmi Pharm. Co. Ltd., 14 Wiryeseong-daero, Songpa-gu, Seoul, 05545, Korea
| | - Matthew D Linnik
- Lilly Biotechnology Center, 10290 Campus Point Drive, San Diego, 92121, USA.
| | - Young-Mi Lee
- Hanmi Research Center, Hanmi Pharm. Co. Ltd., 14 Wiryeseong-daero, Songpa-gu, Seoul, 05545, Korea.
| |
Collapse
|
52
|
Weber MS, Nicholas JA, Yeaman MR. Balancing Potential Benefits and Risks of Bruton Tyrosine Kinase Inhibitor Therapies in Multiple Sclerosis During the COVID-19 Pandemic. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1067. [PMID: 34497100 PMCID: PMC8428017 DOI: 10.1212/nxi.0000000000001067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023]
Abstract
Bruton tyrosine kinase inhibitors (BTKis) encompass a new class of therapeutics currently being evaluated for the treatment of multiple sclerosis (MS). Whether BTKis affect COVID-19 risk or severity or reduce vaccine efficacy are important but unanswered questions. Here, we provide an overview on BTKi mechanisms relevant to COVID-19 infection and vaccination and review preliminary data on BTKi use in patients with COVID-19. BTKis block B-cell receptor– and myeloid fragment crystallizable receptor–mediated signaling, thereby dampening B-cell activation, antibody class-switching, expansion, and cytokine production. Beyond antibodies, COVID-19 severity and vaccine efficacy appear largely linked to T-cell responses and interferon induction, processes not directly affected by BTKis. Given that B cells have clear roles in antigen presentation to T cells, however, it is possible that BTKis may indirectly interfere with beneficial or detrimental T-cell responses during COVID-19 infection or vaccination. In addition to these possible effects on generating a protective immune response, BTKis may attenuate the hyperinflammatory dysregulation often seen in severe cases of COVID-19 that evolves as a key risk factor in this disease. Currently available outcomes from BTKi-treated patients with COVID-19 are discussed. Clinical trials are currently underway to evaluate the safety and efficacy of BTKis in individuals with MS. Although limited data suggest a potential benefit of BTKis on outcomes for some COVID-19 patients, data from adequately powered, prospective and randomized clinical trials are lacking. Likewise, the specific effect of BTKis on the safety and efficacy of COVID-19 vaccines remains to be determined. Any potential unknown risks that BTKi therapy may present to the patient relative to COVID-19 infection, severity, and vaccine efficacy must be balanced with the importance of timely intervention to prevent or minimize MS progression.
Collapse
Affiliation(s)
- Martin S Weber
- From the Institute of Neuropathology (M.S.W.), University Medical Center; Department of Neurology (M.S.W.), University Medical Center, Göttingen, Germany; OhioHealth Multiple Sclerosis Center (J.A.N.), Riverside Methodist Hospital, Columbus, OH; David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles (UCLA); Divisions of Molecular Medicine and Infectious Diseases (M.R.Y.), Harbor-UCLA Medical Center; and Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA.
| | - Jacqueline A Nicholas
- From the Institute of Neuropathology (M.S.W.), University Medical Center; Department of Neurology (M.S.W.), University Medical Center, Göttingen, Germany; OhioHealth Multiple Sclerosis Center (J.A.N.), Riverside Methodist Hospital, Columbus, OH; David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles (UCLA); Divisions of Molecular Medicine and Infectious Diseases (M.R.Y.), Harbor-UCLA Medical Center; and Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| | - Michael R Yeaman
- From the Institute of Neuropathology (M.S.W.), University Medical Center; Department of Neurology (M.S.W.), University Medical Center, Göttingen, Germany; OhioHealth Multiple Sclerosis Center (J.A.N.), Riverside Methodist Hospital, Columbus, OH; David Geffen School of Medicine (M.R.Y.), University of California, Los Angeles (UCLA); Divisions of Molecular Medicine and Infectious Diseases (M.R.Y.), Harbor-UCLA Medical Center; and Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center (M.R.Y.), Torrance, CA
| |
Collapse
|
53
|
Arneson LC, Carroll KJ, Ruderman EM. Bruton's Tyrosine Kinase Inhibition for the Treatment of Rheumatoid Arthritis. Immunotargets Ther 2021; 10:333-342. [PMID: 34485183 PMCID: PMC8409514 DOI: 10.2147/itt.s288550] [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: 06/02/2021] [Accepted: 08/04/2021] [Indexed: 02/02/2023] Open
Abstract
Bruton’s tyrosine kinase (BTK) inhibitors are an emerging class of drugs that inhibit B cell receptor activation, FC-γ receptor signaling, and osteoclast proliferation. Following on approval for treatment of hematologic malignancies, BTK inhibitors are now under investigation to treat a number of different autoimmune diseases, including rheumatoid arthritis (RA). While the results of BTK inhibitors in RA animal models have been promising, the ensuing human clinical trial outcomes have been rather equivocal. This review will outline the mechanisms of BTK inhibition and its potential impact on immune mediated disease, the types of BTK inhibitors being studied for RA, the findings from both preclinical and clinical trials of BTK inhibitors in RA, and directions for future research.
Collapse
Affiliation(s)
- Laura C Arneson
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kristen J Carroll
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eric M Ruderman
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
54
|
Kaul M, End P, Cabanski M, Schuhler C, Jakab A, Kistowska M, Kinhikar A, Maiolica A, Sinn A, Fuhr R, Cenni B. Remibrutinib (LOU064): A selective potent oral BTK inhibitor with promising clinical safety and pharmacodynamics in a randomized phase I trial. Clin Transl Sci 2021; 14:1756-1768. [PMID: 33834628 PMCID: PMC8504815 DOI: 10.1111/cts.13005] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
Safe and effective new oral therapies for autoimmune, allergic, and inflammatory conditions remain a significant therapeutic need. Here, we investigate the human pharmacokinetics, pharmacodynamics (PDs), and safety of the selective, covalent Bruton's tyrosine kinase (BTK) inhibitor, remibrutinib. Study objectives were explored in randomized single and multiple ascending dose (SAD and MAD, respectively) cohorts with daily doses up to 600 mg, and a crossover food effect (FE) cohort, in adult healthy subjects without (SAD [n =80]/FE [n =12]) or with asymptomatic atopic diathesis (MAD [n =64]). A single oral dose of remibrutinib (0.5-600 mg) was rapidly absorbed (time to maximum concentration = 0.5 h-1.25 h) with an apparent blood clearance of 280-560 L/h and apparent volume of distribution of 400-15,000 L. With multiple doses (q.d. and b.i.d.), no pronounced accumulation of remibrutinib was detected (mean residence time was <3 h). Food intake showed no clinically relevant effect on remibrutinib exposure suggesting no need for dose adaptation. With remibrutinib doses greater than or equal to 30 mg, blood BTK occupancy was greater than 95% for at least 24 h (SAD). With MAD, remibrutinib reached near complete blood BTK occupancy at day 12 predose with greater than or equal to 10 mg q.d. Near complete basophil or skin prick test (SPT) inhibition at day 12 predose was achieved at greater than or equal to 50 mg q.d. for CD63 and at greater than or equal to 100 mg q.d. for SPT. Remibrutinib was well-tolerated at all doses without any dose-limiting toxicity. Remibrutinib showed encouraging blood and skin PDs with a favorable safety profile, supporting further development for diseases driven by mast cells, basophils, and B-cells, such as chronic spontaneous urticaria, allergic asthma, or Sjögren's syndrome.
Collapse
Affiliation(s)
- Martin Kaul
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| | - Peter End
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| | | | | | | | | | - Arvind Kinhikar
- Novartis Institutes for Biomedical ResearchCambridgeMassachusettsUSA
| | | | - Angela Sinn
- Early Phase Clinical UnitParexel InternationalBerlinGermany
| | - Rainard Fuhr
- Early Phase Clinical UnitParexel InternationalBerlinGermany
| | - Bruno Cenni
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| |
Collapse
|
55
|
Neys SFH, Rip J, Hendriks RW, Corneth OBJ. Bruton's Tyrosine Kinase Inhibition as an Emerging Therapy in Systemic Autoimmune Disease. Drugs 2021; 81:1605-1626. [PMID: 34609725 PMCID: PMC8491186 DOI: 10.1007/s40265-021-01592-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/14/2022]
Abstract
Systemic autoimmune disorders are complex heterogeneous chronic diseases involving many different immune cells. A significant proportion of patients respond poorly to therapy. In addition, the high burden of adverse effects caused by "classical" anti-rheumatic or immune modulatory drugs provides a need to develop more specific therapies that are better tolerated. Bruton's tyrosine kinase (BTK) is a crucial signaling protein that directly links B-cell receptor (BCR) signals to B-cell activation, proliferation, and survival. BTK is not only expressed in B cells but also in myeloid cells, and is involved in many different signaling pathways that drive autoimmunity. This makes BTK an interesting therapeutic target in the treatment of autoimmune diseases. The past decade has seen the emergence of first-line BTK small-molecule inhibitors with great efficacy in the treatment of B-cell malignancies, but with unfavorable safety profiles for use in autoimmunity due to off-target effects. The development of second-generation BTK inhibitors with superior BTK specificity has facilitated the investigation of their efficacy in clinical trials with autoimmune patients. In this review, we discuss the role of BTK in key signaling pathways involved in autoimmunity and provide an overview of the different inhibitors that are currently being investigated in clinical trials of systemic autoimmune diseases, including rheumatoid arthritis and systemic lupus erythematosus, as well as available results from completed trials.
Collapse
Affiliation(s)
- Stefan F H Neys
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jasper Rip
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| | - Odilia B J Corneth
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
56
|
Abstract
The focus of this review is to examine the role of ITK signaling in multiple diseases and investigate the clinical potential of ITK inhibition. The diseases and potential interventions reviewed include T cell-derived malignancies as well as other neoplastic diseases, allergic diseases such as asthma and atopic dermatitis, certain infectious diseases, several autoimmune disorders such as rheumatoid arthritis and psoriasis, and finally the use of ITK inhibition in both solid organ and bone marrow transplantation recipients.
Collapse
Affiliation(s)
- Samuel Weeks
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 766 Irving Avenue Weiskotten Hall Suite 2281, Syracuse, NY 13210, USA
| | - Rebecca Harris
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 766 Irving Avenue Weiskotten Hall Suite 2281, Syracuse, NY 13210, USA
| | - Mobin Karimi
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 766 Irving Avenue Weiskotten Hall Suite 2281, Syracuse, NY 13210, USA
| |
Collapse
|
57
|
Recent Advances in BTK Inhibitors for the Treatment of Inflammatory and Autoimmune Diseases. Molecules 2021; 26:molecules26164907. [PMID: 34443496 PMCID: PMC8399599 DOI: 10.3390/molecules26164907] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 11/17/2022] Open
Abstract
Bruton’s tyrosine kinase (BTK) plays a crucial role in B-cell receptor and Fc receptor signaling pathways. BTK is also involved in the regulation of Toll-like receptors and chemokine receptors. Given the central role of BTK in immunity, BTK inhibition represents a promising therapeutic approach for the treatment of inflammatory and autoimmune diseases. Great efforts have been made in developing BTK inhibitors for potential clinical applications in inflammatory and autoimmune diseases. This review covers the recent development of BTK inhibitors at preclinical and clinical stages in treating these diseases. Individual examples of three types of inhibitors, namely covalent irreversible inhibitors, covalent reversible inhibitors, and non-covalent reversible inhibitors, are discussed with a focus on their structure, bioactivity and selectivity. Contrary to expectations, reversible BTK inhibitors have not yielded a significant breakthrough so far. The development of covalent, irreversible BTK inhibitors has progressed more rapidly. Many candidates entered different stages of clinical trials; tolebrutinib and evobrutinib are undergoing phase 3 clinical evaluation. Rilzabrutinib, a covalent reversible BTK inhibitor, is now in phase 3 clinical trials and also offers a promising future. An analysis of the protein–inhibitor interactions based on published co-crystal structures provides useful clues for the rational design of safe and effective small-molecule BTK inhibitors.
Collapse
|
58
|
Xing Y, Chu KA, Wadhwa J, Chen W, Zhu J, Bradshaw JM, Shu J, Foulke MC, Loewenstein N, Nunn P, By K, Phiasivongsa P, Goldstein DM, Langrish CL. Preclinical Mechanisms of Topical PRN473, a Bruton Tyrosine Kinase Inhibitor, in Immune-Mediated Skin Disease Models. Immunohorizons 2021; 5:581-589. [PMID: 34326199 DOI: 10.4049/immunohorizons.2100063] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 11/19/2022] Open
Abstract
The expression of Bruton tyrosine kinase (BTK) in B cells and innate immune cells provides essential downstream signaling for BCR, Fc receptors, and other innate immune cell pathways. The topical covalent BTK inhibitor PRN473 has shown durable, reversible BTK occupancy with rapid on-rate and slow off-rate binding kinetics and long residence time, resulting in prolonged, localized efficacy with low systemic exposure in vivo. Mechanisms of PRN473 include inhibition of IgE (FcεR)-mediated activation of mast cells and basophils, IgG (FcγR)-mediated activation of monocytes, and neutrophil migration. In vivo, oral PRN473 was efficacious and well tolerated in the treatment of canine pemphigus foliaceus. In this study, we evaluated in vitro selectivity and functionality, in vivo skin Ab inflammatory responses, and systemic pharmacology with topically administered PRN473. Significant dose-dependent inhibition of IgG-mediated passive Arthus reaction in rats was observed with topical PRN473 and was maintained when given 16 h prior to challenge, reinforcing extended activity with once-daily administration. Similarly, topical PRN473 resulted in significant dose-dependent inhibition of the mouse passive cutaneous anaphylaxis IgE-mediated reaction. Multiday treatment with topical PRN473 in rodents resulted in low-to-no systemic accumulation, suggesting that efficacy was mainly due to localized exposure. Reduced skin Ab inflammatory activity was also confirmed with oral PRN473. These preclinical studies provide a strong biologic basis for targeting innate immune cell responses locally in the skin, with rapid onset of action following once-daily topical PRN473 administration and minimal systemic exposure. Dose-dependent inhibition in these preclinical models of immune-mediated skin diseases support future clinical studies.
Collapse
Affiliation(s)
- Yan Xing
- Principia Biopharma Inc., a Sanofi Company, South San Francisco, CA
| | - Katherine A Chu
- Principia Biopharma Inc., a Sanofi Company, South San Francisco, CA
| | - Jyoti Wadhwa
- Principia Biopharma Inc., a Sanofi Company, South San Francisco, CA
| | - Wei Chen
- Principia Biopharma Inc., a Sanofi Company, South San Francisco, CA
| | - Jiang Zhu
- Principia Biopharma Inc., a Sanofi Company, South San Francisco, CA
| | | | - Jin Shu
- Principia Biopharma Inc., a Sanofi Company, South San Francisco, CA
| | - Matthew C Foulke
- Principia Biopharma Inc., a Sanofi Company, South San Francisco, CA
| | | | - Philip Nunn
- Principia Biopharma Inc., a Sanofi Company, South San Francisco, CA
| | - Kolbot By
- Principia Biopharma Inc., a Sanofi Company, South San Francisco, CA
| | | | | | | |
Collapse
|
59
|
Abstract
Chronic spontaneous urticaria (CSU) is characterized by the presence of wheals, angioedema, or both for at least 6 weeks. It may persist for a long time-up to 50% of the patients have been reported to be symptomatic 5 years after the onset. Some patients can suffer more than one episode of CSU during their lifetime. Considering the recurrences, disabling symptoms, and significant impact on quality of life, proper and effective treatment of CSU is critical. The use of antihistamines (AHs) is still the mainstay of treatment. However, given the low rates of response to AHs (38.6% and 63.2% to standard doses and higher doses, respectively), the complete control of symptoms seems difficult to attain. The use of omalizumab for CSU has been a major breakthrough in the care of patients with CSU. However, the partial response and lack of response to omalizumab in a subgroup of patients, as high as 70% in some studies, make the development of alternative treatments desirable. Ever-increasing knowledge on the pathogenesis is making new target molecules available and enabling drug development for CSU. In addition to drug repurposing as in anti-IL-4/13, IL-5, and IL-17 antibodies, novel targeted therapy options such as ligelizumab and Bruton's tyrosine kinase inhibitors are currently undergoing clinical trials and will be available in the near future. This article reviews the current challenges in the treatment of CSU, the pathogenesis and potential target molecules, and the rationale for novel treatments and their rapidly developing status.
Collapse
|
60
|
Roszkowski L, Ciechomska M. Tuning Monocytes and Macrophages for Personalized Therapy and Diagnostic Challenge in Rheumatoid Arthritis. Cells 2021; 10:cells10081860. [PMID: 34440629 PMCID: PMC8392289 DOI: 10.3390/cells10081860] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/21/2022] Open
Abstract
Monocytes/macrophages play a central role in chronic inflammatory disorders, including rheumatoid arthritis (RA). Activation of these cells results in the production of various mediators responsible for inflammation and RA pathogenesis. On the other hand, the depletion of macrophages using specific antibodies or chemical agents can prevent their synovial tissue infiltration and subsequently attenuates inflammation. Their plasticity is a major feature that helps the switch from a pro-inflammatory phenotype (M1) to an anti-inflammatory state (M2). Therefore, understanding the precise strategy targeting pro-inflammatory monocytes/macrophages should be a powerful way of inhibiting chronic inflammation and bone erosion. In this review, we demonstrate potential consequences of different epigenetic regulations on inflammatory cytokines production by monocytes. In addition, we present unique profiles of monocytes/macrophages contributing to identification of new biomarkers of disease activity or predicting treatment response in RA. We also outline novel approaches of tuning monocytes/macrophages by biologic drugs, small molecules or by other therapeutic modalities to reduce arthritis. Finally, the importance of cellular heterogeneity of monocytes/macrophages is highlighted by single-cell technologies, which leads to the design of cell-specific therapeutic protocols for personalized medicine in RA in the future.
Collapse
|
61
|
Neys SFH, Hendriks RW, Corneth OBJ. Targeting Bruton's Tyrosine Kinase in Inflammatory and Autoimmune Pathologies. Front Cell Dev Biol 2021; 9:668131. [PMID: 34150760 PMCID: PMC8213343 DOI: 10.3389/fcell.2021.668131] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
Bruton's tyrosine kinase (BTK) was discovered due to its importance in B cell development, and it has a critical role in signal transduction downstream of the B cell receptor (BCR). Targeting of BTK with small molecule inhibitors has proven to be efficacious in several B cell malignancies. Interestingly, recent studies reveal increased BTK protein expression in circulating resting B cells of patients with systemic autoimmune disease (AID) compared with healthy controls. Moreover, BTK phosphorylation following BCR stimulation in vitro was enhanced. In addition to its role in BCR signaling, BTK is involved in many other pathways, including pattern recognition, Fc, and chemokine receptor signaling in B cells and myeloid cells. This broad involvement in several immunological pathways provides a rationale for the targeting of BTK in the context of inflammatory and systemic AID. Accordingly, numerous in vitro and in vivo preclinical studies support the potential of BTK targeting in these conditions. Efficacy of BTK inhibitors in various inflammatory and AID has been demonstrated or is currently evaluated in clinical trials. In addition, very recent reports suggest that BTK inhibition may be effective as immunosuppressive therapy to diminish pulmonary hyperinflammation in coronavirus disease 2019 (COVID-19). Here, we review BTK's function in key signaling pathways in B cells and myeloid cells. Further, we discuss recent advances in targeting BTK in inflammatory and autoimmune pathologies.
Collapse
|
62
|
Isenberg D, Furie R, Jones NS, Guibord P, Galanter J, Lee C, McGregor A, Toth B, Rae J, Hwang O, Desai R, Lokku A, Ramamoorthi N, Hackney JA, Miranda P, de Souza VA, Jaller-Raad JJ, Maura Fernandes A, Garcia Salinas R, Chinn LW, Townsend MJ, Morimoto AM, Tuckwell K. Efficacy, Safety, and Pharmacodynamic Effects of the Bruton's Tyrosine Kinase Inhibitor Fenebrutinib (GDC-0853) in Systemic Lupus Erythematosus: Results of a Phase II, Randomized, Double-Blind, Placebo-Controlled Trial. Arthritis Rheumatol 2021; 73:1835-1846. [PMID: 34042314 DOI: 10.1002/art.41811] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/11/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Fenebrutinib (GDC-0853) is a noncovalent, oral, and highly selective inhibitor of Bruton's tyrosine kinase (BTK). The efficacy, safety, and pharmacodynamics of fenebrutinib in systemic lupus erythematosus (SLE) were assessed in this phase II, multicenter, randomized, placebo-controlled study. METHODS Patients who had moderately to severely active SLE while receiving background standard therapy were randomized to receive placebo, fenebrutinib 150 mg once daily, or fenebrutinib 200 mg twice daily. Glucocorticoid taper was recommended from weeks 0 to 12 and from weeks 24 to 36. The primary end point was the SLE Responder Index 4 (SRI-4) response at week 48. RESULTS Patients (n = 260) were enrolled from 44 sites in 12 countries, with the majority from Latin America, the US, and Western Europe. The SRI-4 response rates at week 48 were 51% for fenebrutinib 150 mg once daily (P = 0.37 versus placebo), 52% for fenebrutinib 200 mg twice daily (P = 0.34 versus placebo), and 44% for placebo. British Isles Lupus Assessment Group-based Combined Lupus Assessment response rates at week 48 were 53% for fenebrutinib 150 mg once daily (P = 0.086 versus placebo), 42% for fenebrutinib 200 mg twice daily (P = 0.879 versus placebo), and 41% for placebo. Safety results were similar across all arms, although serious adverse events were more frequent with fenebrutinib 200 mg twice daily. By week 48, patients treated with fenebrutinib had reduced levels of a BTK-dependent plasmablast RNA signature, anti-double-stranded DNA autoantibodies, total IgG, and IgM, as well as increased complement C4 levels, all relative to placebo. CONCLUSION While fenebrutinib had an acceptable safety profile, the primary end point, SRI-4 response, was not met despite evidence of strong pathway inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | - Chin Lee
- Genentech, Inc., South San Francisco, California
| | | | - Balazs Toth
- Genentech, Inc., South San Francisco, California
| | - Julie Rae
- Genentech, Inc., South San Francisco, California
| | - Olivia Hwang
- Genentech, Inc., South San Francisco, California
| | - Rupal Desai
- Genentech, Inc., South San Francisco, California
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Tichenor MS, Wiener JJM, Rao NL, Pooley Deckhut C, Barbay JK, Kreutter KD, Bacani GM, Wei J, Chang L, Murrey HE, Wang W, Ahn K, Huber M, Rex E, Coe KJ, Wu J, Seierstad M, Bembenek SD, Leonard KA, Lebsack AD, Venable JD, Edwards JP. Discovery of a Potent and Selective Covalent Inhibitor of Bruton's Tyrosine Kinase with Oral Anti-Inflammatory Activity. ACS Med Chem Lett 2021; 12:782-790. [PMID: 34055226 DOI: 10.1021/acsmedchemlett.1c00044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/28/2021] [Indexed: 11/28/2022] Open
Abstract
Bruton's tyrosine kinase (BTK) is a cytoplasmic tyrosine kinase that plays a critical role in the activation of B cells, macrophages, and osteoclasts. Given the key role of these cell types in the pathology of autoimmune disorders, BTK inhibitors have the potential to improve treatment outcomes in multiple diseases. Herein, we report the discovery and characterization of a novel potent and selective covalent 4-oxo-4,5-dihydro-3H-1-thia-3,5,8-triazaacenaphthylene-2-carboxamide BTK inhibitor chemotype. Compound 27 irreversibly inhibits BTK by targeting a noncatalytic cysteine residue (Cys481) for covalent bond formation. Compound 27 is characterized by selectivity for BTK, potent in vivo BTK occupancy that is sustained after it is cleared from systemic circulation, and dose-dependent efficacy at reducing joint inflammation in a rat collagen-induced arthritis model.
Collapse
Affiliation(s)
- Mark S. Tichenor
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - John J. M. Wiener
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Navin L. Rao
- Janssen Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477-0776, United States
| | - Charlotte Pooley Deckhut
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - J. Kent Barbay
- Janssen Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477-0776, United States
| | - Kevin D. Kreutter
- Janssen Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477-0776, United States
| | - Genesis M. Bacani
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Jianmei Wei
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Leon Chang
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Heather E. Murrey
- Janssen Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477-0776, United States
| | - Weixue Wang
- Janssen Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477-0776, United States
| | - Kay Ahn
- Janssen Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477-0776, United States
| | - Michael Huber
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Elizabeth Rex
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Kevin J. Coe
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - JieJun Wu
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Mark Seierstad
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Scott D. Bembenek
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Kristi A. Leonard
- Janssen Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477-0776, United States
| | - Alec D. Lebsack
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Jennifer D. Venable
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - James P. Edwards
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| |
Collapse
|
64
|
Fleischmann R. The Results of Well-conducted Negative Clinical Trials Should Be Reported in a Peer-reviewed Journal. J Rheumatol 2021; 48:957-959. [PMID: 33858981 DOI: 10.3899/jrheum.201622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We expect that the pathogenesis, manifestations, and successful management of disease will be fully reported in peer-reviewed journals. However, there are multiple publications addressing the likelihood that clinical trials that do not report a positive result are underreported in the medical literature, with a maximum of 50% of negative studies published, even after 5 years of availability of their results1,2.
Collapse
Affiliation(s)
- Roy Fleischmann
- R. Fleischmann, Clinical Professor of Medicine, MD, Metroplex Clinical Research Center, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
65
|
Estupiñán HY, Berglöf A, Zain R, Smith CIE. Comparative Analysis of BTK Inhibitors and Mechanisms Underlying Adverse Effects. Front Cell Dev Biol 2021; 9:630942. [PMID: 33777941 PMCID: PMC7991787 DOI: 10.3389/fcell.2021.630942] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/03/2021] [Indexed: 12/16/2022] Open
Abstract
The cytoplasmic protein-tyrosine kinase BTK plays an essential role for differentiation and survival of B-lineage cells and, hence, represents a suitable drug target. The number of BTK inhibitors (BTKis) in the clinic has increased considerably and currently amounts to at least 22. First-in-class was ibrutinib, an irreversible binder forming a covalent bond to a cysteine in the catalytic region of the kinase, for which we have identified 228 active trials listed at ClinicalTrials.gov. Next-generation inhibitors, acalabrutinib and zanubrutinib, are approved both in the United States and in Europe, and zanubrutinib also in China, while tirabrutinib is currently only registered in Japan. In most cases, these compounds have been used for the treatment of B-lymphocyte tumors. However, an increasing number of trials instead addresses autoimmunity and inflammation in multiple sclerosis, rheumatoid arthritis, pemphigus and systemic lupus erythematosus with the use of either irreversibly binding inhibitors, e.g., evobrutinib and tolebrutinib, or reversibly binding inhibitors, like fenebrutinib. Adverse effects (AEs) have predominantly implicated inhibition of other kinases with a BTKi-binding cysteine in their catalytic domain. Analysis of the reported AEs suggests that ibrutinib-associated atrial fibrillation is caused by binding to ERBB2/HER2 and ERBB4/HER4. However, the binding pattern of BTKis to various additional kinases does not correlate with the common assumption that skin manifestations and diarrhoeas are off-target effects related to EGF receptor inhibition. Moreover, dermatological toxicities, diarrhoea, bleedings and invasive fungal infections often develop early after BTKi treatment initiation and subsequently subside. Conversely, cardiovascular AEs, like hypertension and various forms of heart disease, often persist.
Collapse
Affiliation(s)
- H. Yesid Estupiñán
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
- Departamento de Ciencias Básicas, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Anna Berglöf
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Rula Zain
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
- Centre for Rare Diseases, Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - C. I. Edvard Smith
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
66
|
Liu J, Ting JP, Al-Azzam S, Ding Y, Afshar S. Therapeutic Advances in Diabetes, Autoimmune, and Neurological Diseases. Int J Mol Sci 2021; 22:ijms22062805. [PMID: 33802091 PMCID: PMC8001105 DOI: 10.3390/ijms22062805] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 02/08/2023] Open
Abstract
Since 2015, 170 small molecules, 60 antibody-based entities, 12 peptides, and 15 gene- or cell-therapies have been approved by FDA for diverse disease indications. Recent advancement in medicine is facilitated by identification of new targets and mechanisms of actions, advancement in discovery and development platforms, and the emergence of novel technologies. Early disease detection, precision intervention, and personalized treatments have revolutionized patient care in the last decade. In this review, we provide a comprehensive overview of current and emerging therapeutic modalities developed in the recent years. We focus on nine diseases in three major therapeutics areas, diabetes, autoimmune, and neurological disorders. The pathogenesis of each disease at physiological and molecular levels is discussed and recently approved drugs as well as drugs in the clinic are presented.
Collapse
Affiliation(s)
- Jinsha Liu
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Joey Paolo Ting
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Shams Al-Azzam
- Professional Scientific Services, Eurofins Lancaster Laboratories, Lancaster, PA 17605, USA;
| | - Yun Ding
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Sepideh Afshar
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
- Correspondence:
| |
Collapse
|
67
|
von Hundelshausen P, Siess W. Bleeding by Bruton Tyrosine Kinase-Inhibitors: Dependency on Drug Type and Disease. Cancers (Basel) 2021; 13:1103. [PMID: 33806595 PMCID: PMC7961939 DOI: 10.3390/cancers13051103] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Bruton tyrosine kinase (Btk) is expressed in B-lymphocytes, myeloid cells and platelets, and Btk-inhibitors (BTKi) are used to treat patients with B-cell malignancies, developed against autoimmune diseases, have been proposed as novel antithrombotic drugs, and been tested in patients with severe COVID-19. However, mild bleeding is frequent in patients with B-cell malignancies treated with the irreversible BTKi ibrutinib and the recently approved 2nd generation BTKi acalabrutinib, zanubrutinib and tirabrutinib, and also in volunteers receiving in a phase-1 study the novel irreversible BTKi BI-705564. In contrast, no bleeding has been reported in clinical trials of other BTKi. These include the brain-penetrant irreversible tolebrutinib and evobrutinib (against multiple sclerosis), the irreversible branebrutinib, the reversible BMS-986142 and fenebrutinib (targeting rheumatoid arthritis and lupus erythematodes), and the reversible covalent rilzabrutinib (against pemphigus and immune thrombocytopenia). Remibrutinib, a novel highly selective covalent BTKi, is currently in clinical studies of autoimmune dermatological disorders. This review describes twelve BTKi approved or in clinical trials. By focusing on their pharmacological properties, targeted disease, bleeding side effects and actions on platelets it attempts to clarify the mechanisms underlying bleeding. Specific platelet function tests in blood might help to estimate the probability of bleeding of newly developed BTKi.
Collapse
Affiliation(s)
- Philipp von Hundelshausen
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University (LMU), 80336 Munich, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Wolfgang Siess
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University (LMU), 80336 Munich, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
68
|
Fenebrutinib in H 1 antihistamine-refractory chronic spontaneous urticaria: a randomized phase 2 trial. Nat Med 2021; 27:1961-1969. [PMID: 34750553 PMCID: PMC8604722 DOI: 10.1038/s41591-021-01537-w] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 09/10/2021] [Indexed: 12/28/2022]
Abstract
Bruton's tyrosine kinase (BTK) is crucial for FcεRI-mediated mast cell activation and essential for autoantibody production by B cells in chronic spontaneous urticaria (CSU). Fenebrutinib, an orally administered, potent, highly selective, reversible BTK inhibitor, may be effective in CSU. This double-blind, placebo-controlled, phase 2 trial (EudraCT ID 2016-004624-35 ) randomized 93 adults with antihistamine-refractory CSU to 50 mg daily, 150 mg daily and 200 mg twice daily of fenebrutinib or placebo for 8 weeks. The primary end point was change from baseline in urticaria activity score over 7 d (UAS7) at week 8. Secondary end points were the change from baseline in UAS7 at week 4 and the proportion of patients well-controlled (UAS7 ≤ 6) at week 8. Fenebrutinib efficacy in patients with type IIb autoimmunity and effects on IgG-anti-FcεRI were exploratory end points. Safety was also evaluated. The primary end point was met, with dose-dependent improvements in UAS7 at week 8 occurring at 200 mg twice daily and 150 mg daily, but not at 50 mg daily of fenebrutinib versus placebo. Asymptomatic, reversible grade 2 and 3 liver transaminase elevations occurred in the fenebrutinib 150 mg daily and 200 mg twice daily groups (2 patients each). Fenebrutinib diminished disease activity in patients with antihistamine-refractory CSU, including more patients with refractory type IIb autoimmunity. These results support the potential use of BTK inhibition in antihistamine-refractory CSU.
Collapse
|
69
|
Bilal M, Qindeel M, Nunes LV, Duarte MTS, Ferreira LFR, Soriano RN, Iqbal HMN. Marine-Derived Biologically Active Compounds for the Potential Treatment of Rheumatoid Arthritis. Mar Drugs 2020; 19:10. [PMID: 33383638 PMCID: PMC7823916 DOI: 10.3390/md19010010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/05/2020] [Accepted: 12/10/2020] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease with a prevalence rate of up to 1% and is significantly considered a common worldwide public health concern. Commercially, several traditional formulations are available to treat RA to some extent. However, these synthetic compounds exert toxicity and considerable side effects even at lower therapeutic concentrations. Considering the above-mentioned critiques, research is underway around the world in finding and exploiting potential alternatives. For instance, marine-derived biologically active compounds have gained much interest and are thus being extensively utilized to confront the confines of in practice counterparts, which have become ineffective for 21st-century medical settings. The utilization of naturally available bioactive compounds and their derivatives can minimize these synthetic compounds' problems to treat RA. Several marine-derived compounds exhibit anti-inflammatory and antioxidant properties and can be effectively used for therapeutic purposes against RA. The results of several studies ensured that the extraction of biologically active compounds from marine sources could provide a new and safe source for drug development against RA. Finally, current challenges, gaps, and future perspectives have been included in this review.
Collapse
Affiliation(s)
- Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China
| | - Maimoona Qindeel
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Leonardo Vieira Nunes
- Department of Medicine, Federal University of Juiz de Fora, Juiz de Fora-MG 36036-900, Brazil;
| | | | - Luiz Fernando Romanholo Ferreira
- Graduate Program in Process Engineering, Tiradentes University (UNIT), Av. Murilo Dantas, 300, Farolândia, Aracaju-Sergipe 49032-490, Brazil;
- Institute of Technology and Research (ITP), Tiradentes University (UNIT), Av. Murilo Dantas, 300, Farolândia, Aracaju-Sergipe 49032-490, Brazil
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares-MG 35010-180, Brazil;
| | - Hafiz M. N. Iqbal
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico
| |
Collapse
|
70
|
Schwaid AG, Spencer KB. Strategies for Targeting the NLRP3 Inflammasome in the Clinical and Preclinical Space. J Med Chem 2020; 64:101-122. [PMID: 33351619 DOI: 10.1021/acs.jmedchem.0c01307] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inhibiting the NLRP3 inflammasome mediates inflammation in an extensive number of preclinical models. As excitement in this field has grown, several companies have recently initiated testing of direct NLRP3 inhibitors in the clinic. At the same time, the NLRP3 inflammasome is part of a larger pro-inflammatory pathway, whose modulation is also being explored. Multiple targets in this pathway are already impinged upon by molecules that have been through clinical trials. These data, informed by the growing mechanistic understanding of the NLRP3 inflammasome in the preclinical space, provide a rich backdrop to assess the current state of the field. Here we explore attempts to inhibit the NLRP3 inflammasome in light of clinical and preclinical data around efficacy and safety.
Collapse
Affiliation(s)
- Adam G Schwaid
- Chemical Biology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Kerrie B Spencer
- Chemical Biology, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| |
Collapse
|
71
|
Genovese MC, Spindler A, Sagawa A, Park W, Dudek A, Kivitz A, Chao J, Chan LSM, Witcher J, Barchuk W, Nirula A. Safety and Efficacy of Poseltinib, Bruton's Tyrosine Kinase Inhibitor, in Patients With Rheumatoid Arthritis: A Randomized, Double-blind, Placebo-controlled, 2-part Phase II Study. J Rheumatol 2020; 48:969-976. [PMID: 33323529 DOI: 10.3899/jrheum.200893] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To evaluate the efficacy and safety of poseltinib (formerly LY3337641/HM71224), an irreversible covalent inhibitor of Bruton's tyrosine kinase in a 2-part, phase II trial (RAjuvenate; ClinicalTrials.gov: NCT02628028) in adults with active rheumatoid arthritis (RA). METHODS In Part A, 36 patients with mildly active RA were randomized 1:1:1:1 to oral poseltinib 5, 10, or 30 mg or placebo once daily for 4 weeks to assess safety and tolerability. No safety signals precluded moving to Part B, where 250 patients with moderate-to-severe RA were randomized 1:1:1:1 to oral poseltinib 5 mg (n = 63), 10 mg (n = 62), or 30 mg (n = 63), or placebo (n = 62) once daily for 12 weeks. Parts A and B permitted stable doses of background disease-modifying antirheumatic drugs. The primary endpoint in Part B was proportion of patients achieving 20% improvement in American College of Rheumatology criteria (ACR20) at Week 12. Logistic regression compared each poseltinib dose to placebo for primary and secondary endpoints. Nonresponder imputation was used for missing data. RESULTS After interim analysis showed low likelihood of demonstrating significant efficacy, the sponsor discontinued Part B of the study. One hundred and eighty-nine (76%) patients completed 12 weeks in Part B; 61 discontinued study treatment (27 [44%] due to study termination by sponsor). There was no statistically significant difference in ACR20 response between any dose of poseltinib and placebo at Week 12 (P > 0.05 for all comparisons). Five serious adverse events occurred (n = 2, placebo; n = 3, 30 mg); there was 1 death due to a fall. CONCLUSION While no safety findings precluded continuation, the study was terminated after interim data demonstrated low likelihood of benefit in RA.
Collapse
Affiliation(s)
- Mark C Genovese
- M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University, Palo Alto, California, USA;
| | - Alberto Spindler
- A. Spindler, MD, Centro Medico Privado de Reumatologia, Tucumán, Argentina
| | - Akira Sagawa
- A. Sagawa, MD, Sagawa Akira Rheumatology Clinic, Sapporo, Japan
| | - Won Park
- W. Park, MD, PhD, Division of Rheumatology, IN-HA University, Incheon, Korea
| | - Anna Dudek
- A. Dudek, MD, PhD, AMED Medical Center, Warsaw, Poland
| | - Alan Kivitz
- A. Kivitz MD, CPI, Altoona Center for Clinical Research, Duncansville, Pennsylvania, USA
| | - Jeannie Chao
- J. Chao, MD, L. Chan, MS, J. Witcher, PhD, W. Barchuk, MD, A. Nirula, MD, PhD, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Lai Shan Melanie Chan
- J. Chao, MD, L. Chan, MS, J. Witcher, PhD, W. Barchuk, MD, A. Nirula, MD, PhD, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Jennifer Witcher
- J. Chao, MD, L. Chan, MS, J. Witcher, PhD, W. Barchuk, MD, A. Nirula, MD, PhD, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - William Barchuk
- J. Chao, MD, L. Chan, MS, J. Witcher, PhD, W. Barchuk, MD, A. Nirula, MD, PhD, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Ajay Nirula
- J. Chao, MD, L. Chan, MS, J. Witcher, PhD, W. Barchuk, MD, A. Nirula, MD, PhD, Eli Lilly and Company, Indianapolis, Indiana, USA
| |
Collapse
|
72
|
Harbi MH, Smith CW, Nicolson PLR, Watson SP, Thomas MR. Novel antiplatelet strategies targeting GPVI, CLEC-2 and tyrosine kinases. Platelets 2020; 32:29-41. [PMID: 33307909 DOI: 10.1080/09537104.2020.1849600] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Antiplatelet medications comprise the cornerstone of treatment for diseases that involve arterial thrombosis, including acute coronary syndromes (ACS), stroke and peripheral arterial disease. However, antiplatelet medications may cause bleeding and, furthermore, thrombotic events may still recur despite treatment. The interaction of collagen with GPVI receptors on the surface of platelets has been identified as one of the major players in the pathophysiology of arterial thrombosis that occurs following atherosclerotic plaque rupture. Promisingly, GPVI deficiency in humans appears to have a minimal impact on bleeding. These findings together suggest that targeting platelet GPVI may provide a novel treatment strategy that provides additional antithrombotic efficacy with minimal disruption of normal hemostasis compared to conventional antiplatelet medications. CLEC-2 is gaining interest as a therapeutic target for a variety of thrombo-inflammatory disorders including deep vein thrombosis (DVT) with treatment also predicted to cause minimal disruption to hemostasis. GPVI and CLEC-2 signal through Src, Syk and Tec family tyrosine kinases, providing additional strategies for inhibiting both receptors. In this review, we summarize the evidence regarding GPVI and CLEC-2 and strategies for inhibiting these receptors to inhibit platelet recruitment and activation in thrombotic diseases.
Collapse
Affiliation(s)
- Maan H Harbi
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Christopher W Smith
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Phillip L R Nicolson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,University Hospitals Birmingham NHS Foundation Trust , Birmingham, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,University Hospitals Birmingham NHS Foundation Trust , Birmingham, UK.,Sandwell and West Birmingham NHS Trust , Birmingham, UK
| |
Collapse
|
73
|
Srivastava AS, Ko S, Watterson SH, Pattoli MA, Skala S, Cheng L, Obermeier MT, Vickery R, Discenza LN, D’Arienzo CJ, Gillooly KM, Taylor TL, Pulicicchio C, McIntyre KW, Yip S, Li P, Sun D, Wu DR, Dai J, Wang C, Zhang Y, Wang B, Pawluczyk J, Kempson J, Zhao R, Hou X, Rampulla R, Mathur A, Galella MA, Salter-Cid L, Barrish JC, Carter PH, Fura A, Burke JR, Tino JA. Driving Potency with Rotationally Stable Atropisomers: Discovery of Pyridopyrimidinedione-Carbazole Inhibitors of BTK. ACS Med Chem Lett 2020; 11:2195-2203. [PMID: 33214829 DOI: 10.1021/acsmedchemlett.0c00335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/16/2020] [Indexed: 12/25/2022] Open
Abstract
Bruton's tyrosine kinase (BTK) has been shown to play a key role in the pathogenesis of autoimmunity. Therefore, the inhibition of the kinase activity of BTK with a small molecule inhibitor could offer a breakthrough in the clinical treatment of many autoimmune diseases. This Letter describes the discovery of BMS-986143 through systematic structure-activity relationship (SAR) development. This compound benefits from defined chirality derived from two rotationally stable atropisomeric axes, providing a potent and selective single atropisomer with desirable efficacy and tolerability profiles.
Collapse
Affiliation(s)
- Anurag S. Srivastava
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Soo Ko
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Scott H. Watterson
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Mark A. Pattoli
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Stacey Skala
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Lihong Cheng
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Mary T. Obermeier
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Rodney Vickery
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Lorell N. Discenza
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Celia J. D’Arienzo
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Kathleen M. Gillooly
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Tracy L. Taylor
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Claudine Pulicicchio
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Kim W. McIntyre
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Shiuhang Yip
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Peng Li
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Dawn Sun
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Dauh-Rurng Wu
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Jun Dai
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Chunlei Wang
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Yingru Zhang
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Bei Wang
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Joseph Pawluczyk
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - James Kempson
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Rulin Zhao
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Xiaoping Hou
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Richard Rampulla
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Arvind Mathur
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Michael A. Galella
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Luisa Salter-Cid
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Joel C. Barrish
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Percy H. Carter
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Aberra Fura
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - James R. Burke
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Joseph A. Tino
- Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| |
Collapse
|
74
|
Siess W, Hundelshausen PV, Lorenz R. Selective inhibition of thromboinflammation in COVID-19 by Btk inhibitors. Platelets 2020; 31:989-992. [PMID: 32892684 DOI: 10.1080/09537104.2020.1809647] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Wolfgang Siess
- Institute for Prevention of Cardiovascular Diseases, Ludwig-Maximilians University (LMU) , Munich, Germany
| | - Philipp Von Hundelshausen
- Institute for Prevention of Cardiovascular Diseases, Ludwig-Maximilians University (LMU) , Munich, Germany
| | - Reinhard Lorenz
- Institute for Prevention of Cardiovascular Diseases, Ludwig-Maximilians University (LMU) , Munich, Germany
| |
Collapse
|
75
|
Nicolson PL, Welsh JD, Chauhan A, Thomas MR, Kahn ML, Watson SP. A rationale for blocking thromboinflammation in COVID-19 with Btk inhibitors. Platelets 2020; 31:685-690. [PMID: 32552307 DOI: 10.1080/09537104.2020.1775189] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Phillip Lr Nicolson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - John D Welsh
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania , Philadelphia, Pennsylvania, USA
| | - Abhishek Chauhan
- Centre for Liver Research, Institute of Immunology and Inflammation, and National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania , Philadelphia, Pennsylvania, USA
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham , The Midlands, UK
| |
Collapse
|