1
|
Dechenne J, Wierzbicka M, Krimou R, El Aakchioui A, Malo Pueyo J, Messens J, Fillet M, Spillier Q, Frédérick R. Examining Arginase-1 Trimerization Uncovers a Promising Allosteric Site for Inhibition. J Med Chem 2025. [PMID: 39748145 DOI: 10.1021/acs.jmedchem.4c01993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Arginase-1 (ARG-1) is a promising target for cancer immunotherapy, but the small size and the highly polar nature of its catalytic site present significant challenges for inhibitor development. An alternative strategy to induce enzyme inhibition by targeting protein oligomerization has been developed recently, offering several advantages such as increased selectivity, promotion of protein degradation, and potential substoichiometric inhibition. In this study, we demonstrated that only trimeric ARG-1 is active, which was confirmed by producing monomeric arginase-1. Through in silico-driven site-directed mutagenesis, we identified an allosteric site involving five key amino acids responsible for ARG-1 trimerization. We further demonstrated the covalent modification of a key arginine residue within this pocket using phenylglyoxal disrupted ARG-1 oligomerization. Although phenylglyoxal has limited potency, it effectively supports the concept of ARG-1 inhibition via homomeric disruption, validating this allosteric targeting approach.
Collapse
Affiliation(s)
- Juhans Dechenne
- Louvain Drug Research Institute (LDRI), Medicinal Chemistry Research Group (CMFA), Université Catholique de Louvain (UCLouvain), Brussels B-1200, Belgium
| | - Magdalena Wierzbicka
- Laboratory for the Analysis of Medicines (CIRM), Université de Liège (ULG), Liège B-4000, Belgium
| | - Reda Krimou
- Louvain Drug Research Institute (LDRI), Medicinal Chemistry Research Group (CMFA), Université Catholique de Louvain (UCLouvain), Brussels B-1200, Belgium
| | - Asia El Aakchioui
- Louvain Drug Research Institute (LDRI), Medicinal Chemistry Research Group (CMFA), Université Catholique de Louvain (UCLouvain), Brussels B-1200, Belgium
| | - Julia Malo Pueyo
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, Brussels B-1050, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, Brussels B-1050, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels B-1050, Belgium
| | - Joris Messens
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, Brussels B-1050, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, Brussels B-1050, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels B-1050, Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines (CIRM), Université de Liège (ULG), Liège B-4000, Belgium
| | - Quentin Spillier
- Louvain Drug Research Institute (LDRI), Medicinal Chemistry Research Group (CMFA), Université Catholique de Louvain (UCLouvain), Brussels B-1200, Belgium
| | - Raphaël Frédérick
- Louvain Drug Research Institute (LDRI), Medicinal Chemistry Research Group (CMFA), Université Catholique de Louvain (UCLouvain), Brussels B-1200, Belgium
| |
Collapse
|
2
|
Kim MS, Kang H, Baek JH, Cho MG, Chung EJ, Kim SJ, Chung JY, Chun KH. Disrupting Notch signaling related HES1 in myeloid cells reinvigorates antitumor T cell responses. Exp Hematol Oncol 2024; 13:122. [PMID: 39702544 DOI: 10.1186/s40164-024-00588-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are immunosuppressive cells within the tumor microenvironment (TME) that hinder anti-tumor immunity. Notch signaling is a pathway crucial for TAM differentiation and function. Here, we investigate the role of HES1, a downstream target of Notch signaling, in TAM-mediated immunosuppression and explore its potential as a target for cancer immunotherapy. METHODS In this work, we constructed conditional Hes1 knockout mice to selectively delete Hes1 in TAMs. We further analyzed the TME composition, T cell infiltration and activation, and anti-tumor effects in these mice, both alone and in combination with PD-1 checkpoint blockade. RESULTS Our study showed that expression levels of Notch target Hes1 were increase in TAMs and mice with conditional knockout of Hes1 gene in TAMs exhibited decreased tumor growth, with increased infiltration and activation of cytotoxic T cells in tumors. Expression of tumor promoting factors was critically altered in Hes1-conditional KO TAMs, leading to the improved tumor microenvironment. Notably, arginase-1 expression was decreased in Hes1-conditional KO mice. Arg1 is known to deplete arginine and deactivate T cells in the TME. Administration of anti-PD-1 monoclonal antibody inhibited tumor growth to a greater extent in Hes1-conditional KO mice than in WT mice. CONCLUSIONS We identified a pivotal role for the Notch signaling pathway in shaping TAM function, suggesting that T-cell dysfunction in the TME is caused when the Notch target, HES1, in TAMs is upregulated by tumor-associated factors (TAFs), which, in turn, increases the expression of arginase-1. Targeting HES1 in TAMs appears to be a promising strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Myung Sup Kim
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyeokgu Kang
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jung-Hwan Baek
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Moon-Gyu Cho
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Eun Joo Chung
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Seok-Jun Kim
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea
- Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-Associated Disorder Control Technology, Chosun University, Gwangju, 61452, Republic of Korea
- Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju, 61452, Republic of Korea
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Affiliate Faculty, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.
| |
Collapse
|
3
|
Zha C, Yang X, Yang J, Zhang Y, Huang R. Immunosuppressive microenvironment in acute myeloid leukemia: overview, therapeutic targets and corresponding strategies. Ann Hematol 2024:10.1007/s00277-024-06117-9. [PMID: 39607487 DOI: 10.1007/s00277-024-06117-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Similar to other malignancies, immune dysregulation is a key feature of acute myeloid leukemia (AML), manifesting as suppressed anti-leukemia immune cells, immune evasion by leukemia blasts, and disease progression. Various immunosuppressive factors within the AML microenvironment contribute to the weakening of host immune responses and the efficacy of cellular immunotherapy. To address these challenges, strategies targeting immunosuppressive elements within the AML microenvironment aim to bolster host or adoptive immune effector cells, ultimately enhancing leukemia treatment. Additionally, the off-target effects of certain targeted drugs (venetoclax, sorafenib, ivosidenib, etc.) may also positively impact anti-AML immunity and immunotherapy. This review provides an overview of the immunosuppressive factors present in AML microenvironment and the strategies developed to rescue immune cells from immunosuppression. We also outline how targeted agents can alter the immune landscape in AML patients, and discuss the potential of targeted drugs to benefit host anti-leukemia immunity and immunotherapy for AML.
Collapse
Affiliation(s)
- Chenyu Zha
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinyu Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yujie Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Rui Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
4
|
Failla M, Molaro MC, Schiano ME, Serafini M, Tiburtini GA, Gianquinto E, Scoccia R, Battisegola C, Rimoli MG, Chegaev K, Ercolano G, Lazzarato L, Spyrakis F, Sodano F. Opportunities and Challenges of Arginase Inhibitors in Cancer: A Medicinal Chemistry Perspective. J Med Chem 2024; 67:19988-20021. [PMID: 39558532 DOI: 10.1021/acs.jmedchem.4c01429] [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: 11/20/2024]
Abstract
The overexpression of two arginase (ARG) isoforms, ARG1 and ARG2, contributes to the onset of numerous disorders, including cardiovascular and immune-mediated diseases, as well as tumors. To elucidate the specific roles of ARG1 and ARG2 without interfering with their physiological functions, it is crucial to develop effective ARG inhibitors that target only one isoform, while maintaining low toxicity and an adequate pharmacokinetic profile. In this context, we present a comprehensive overview of the different generations of ARG inhibitors. Given the general lack of selectivity in most existing inhibitors, we analyzed the structural features and plasticity of the ARG1 and ARG2 binding sites to explore the potential for designing inhibitors with novel binding patterns. We also review ongoing preclinical and clinical studies on selected inhibitors, highlighting both progress and challenges in developing potent, selective ARG inhibitors. Furthermore, we discuss medicinal chemistry strategies that may accelerate the discovery of selective ARG inhibitors.
Collapse
Affiliation(s)
- Mariacristina Failla
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | | | | | - Marta Serafini
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | | | - Eleonora Gianquinto
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Riccardo Scoccia
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Chiara Battisegola
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Maria Grazia Rimoli
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Giuseppe Ercolano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Francesca Spyrakis
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Federica Sodano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
5
|
Canè S, Geiger R, Bronte V. The roles of arginases and arginine in immunity. Nat Rev Immunol 2024:10.1038/s41577-024-01098-2. [PMID: 39420221 DOI: 10.1038/s41577-024-01098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
Arginase activity and arginine metabolism in immune cells have important consequences for health and disease. Their dysregulation is commonly observed in cancer, autoimmune disorders and infectious diseases. Following the initial description of a role for arginase in the dysfunction of T cells mounting an antitumour response, numerous studies have broadened our understanding of the regulation and expression of arginases and their integration with other metabolic pathways. Here, we highlight the differences in arginase compartmentalization and storage between humans and rodents that should be taken into consideration when assessing the effects of arginase activity. We detail the roles of arginases, arginine and its metabolites in immune cells and their effects in the context of cancer, autoimmunity and infectious disease. Finally, we explore potential therapeutic strategies targeting arginases and arginine.
Collapse
Affiliation(s)
- Stefania Canè
- The Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Roger Geiger
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Università della Svizzera italiana, Bellinzona, Switzerland
| | | |
Collapse
|
6
|
Marzęta-Assas P, Jacenik D, Zasłona Z. Pathophysiology of Arginases in Cancer and Efforts in Their Pharmacological Inhibition. Int J Mol Sci 2024; 25:9782. [PMID: 39337272 PMCID: PMC11431790 DOI: 10.3390/ijms25189782] [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: 08/05/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Arginases are key enzymes that hydrolyze L-arginine to urea and L-ornithine in the urea cycle. The two arginase isoforms, arginase 1 (ARG1) and arginase 2 (ARG2), regulate the proliferation of cancer cells, migration, and apoptosis; affect immunosuppression; and promote the synthesis of polyamines, leading to the development of cancer. Arginases also compete with nitric oxide synthase (NOS) for L-arginine, and their participation has also been confirmed in cardiovascular diseases, stroke, and inflammation. Due to the fact that arginases play a crucial role in the development of various types of diseases, finding an appropriate candidate to inhibit the activity of these enzymes would be beneficial for the therapy of many human diseases. In this review, based on numerous experimental, preclinical, and clinical studies, we provide a comprehensive overview of the biological and physiological functions of ARG1 and ARG2, their molecular mechanisms of action, and affected metabolic pathways. We summarize the recent clinical trials' advances in targeting arginases and describe potential future drugs.
Collapse
Affiliation(s)
| | - Damian Jacenik
- Molecure S.A., 101 Żwirki i Wigury St., 02-089 Warsaw, Poland
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska St., 90-236 Lodz, Poland
| | | |
Collapse
|
7
|
Chattopadhyay S, Hazra R, Mallick A, Gayen S, Roy S. Small-molecule in cancer immunotherapy: Revolutionizing cancer treatment with transformative, game-changing breakthroughs. Biochim Biophys Acta Rev Cancer 2024; 1879:189170. [PMID: 39127244 DOI: 10.1016/j.bbcan.2024.189170] [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: 06/13/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Immunotherapy has revolutionized cancer management, with antibody-based treatments leading the charge due to their superior pharmacodynamics, including enhanced effectiveness and specificity. However, these therapies are hampered by limitations such as prolonged half-lives, poor tissue and tumor penetration, and minimal oral bioavailability. Additionally, their immunogenic nature can cause adverse effects. Consequently, the focus is shifting towards small-molecule-based immunotherapies, which potentially overcome these drawbacks. Emerging as a promising alternative, small molecules offer the benefits of therapeutic antibodies and immunomodulators, often yielding synergistic effects when combined. Recent advancements in small-molecule cancer immunotherapy are notable, featuring inhibitors, agonists, and degraders that act as immunomodulators. This article delves into the current landscape of small-molecule immunotherapy in cancer treatment, highlighting novel agents targeting key pathways such as Toll-like receptors (TLR), PD-1/PD-L1, chemokine receptors, and stimulators of interferon genes (STING). The review emphasizes newly discovered molecular entities and their modulatory roles in tumorigenesis, many of which have progressed to clinical trials, that aims to provide a comprehensive snapshot of the evolving frontier in cancer treatment, driven by small-molecule immunomodulators.
Collapse
Affiliation(s)
- Soumyadeep Chattopadhyay
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Rudradeep Hazra
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Arijit Mallick
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Sakuntala Gayen
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, Kolkata, West Bengal 700053, India.
| |
Collapse
|
8
|
Grobben Y. Targeting amino acid-metabolizing enzymes for cancer immunotherapy. Front Immunol 2024; 15:1440269. [PMID: 39211039 PMCID: PMC11359565 DOI: 10.3389/fimmu.2024.1440269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Despite the immune system's role in the detection and eradication of abnormal cells, cancer cells often evade elimination by exploitation of various immune escape mechanisms. Among these mechanisms is the ability of cancer cells to upregulate amino acid-metabolizing enzymes, or to induce these enzymes in tumor-infiltrating immunosuppressive cells. Amino acids are fundamental cellular nutrients required for a variety of physiological processes, and their inadequacy can severely impact immune cell function. Amino acid-derived metabolites can additionally dampen the anti-tumor immune response by means of their immunosuppressive activities, whilst some can also promote tumor growth directly. Based on their evident role in tumor immune escape, the amino acid-metabolizing enzymes glutaminase 1 (GLS1), arginase 1 (ARG1), inducible nitric oxide synthase (iNOS), indoleamine 2,3-dioxygenase 1 (IDO1), tryptophan 2,3-dioxygenase (TDO) and interleukin 4 induced 1 (IL4I1) each serve as a promising target for immunotherapeutic intervention. This review summarizes and discusses the involvement of these enzymes in cancer, their effect on the anti-tumor immune response and the recent progress made in the preclinical and clinical evaluation of inhibitors targeting these enzymes.
Collapse
|
9
|
Gzik A, Borek B, Chrzanowski J, Jedrzejczak K, Dziegielewski M, Brzezinska J, Nowicka J, Grzybowski MM, Rejczak T, Niedzialek D, Wieczorek G, Olczak J, Golebiowski A, Zaslona Z, Blaszczyk R. Novel orally bioavailable piperidine derivatives as extracellular arginase inhibitors developed by a ring expansion. Eur J Med Chem 2024; 264:116033. [PMID: 38096651 DOI: 10.1016/j.ejmech.2023.116033] [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: 10/24/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
Arginase is a multifaced enzyme that plays an important role in health and disease being regarded as a therapeutic target for the treatment of various pathological states such as malignancies, asthma, and cardiovascular disease. The discovery of boronic acid-based arginase inhibitors in 1997 revolutionized attempts of medicinal chemistry focused on development of drugs targeting arginase. Unfortunately, these very polar compounds had limitations such as analysis and purification without chromophores, synthetically challenging space, and poor oral bioavailability. Herein, we present a novel class of boronic acid-based arginase inhibitors which are piperidine derivatives exhibiting a different pharmacological profile compared to our drug candidate in cancer immunotherapy -OATD-02 - dual ARG1/2 inhibitor with high intracellular activity. Compounds from this new series show low intracellular activity, hence they can inhibit mainly extracellular arginase, providing different therapeutic space compared to a dual intracellular ARG1/2 inhibitor. The disclosed series showed good inhibitory potential towards arginase enzyme in vitro (IC50 up to 160 nM), favorable pharmacokinetics in animal models, and encouraging preliminary in vitro and in vivo tolerability. Compounds from the new series have moderate-to-high oral bioavailability (up to 66 %) and moderate clearance in vivo. Herein we describe the development and optimization of the synthesis of the new class of boronic acid-based arginase inhibitors via a ring expansion approach starting from the inexpensive chirality source (d-hydroxyproline). This upgraded methodology facilitated a gram-scale delivery of the final compound and eliminated the need for costly and time-consuming chiral resolution.
Collapse
Affiliation(s)
- Anna Gzik
- Molecure S.A., Zwirki i Wigury 101, Warsaw, 02-089, Poland
| | | | | | | | | | | | - Julita Nowicka
- Molecure S.A., Zwirki i Wigury 101, Warsaw, 02-089, Poland
| | | | - Tomasz Rejczak
- Molecure S.A., Zwirki i Wigury 101, Warsaw, 02-089, Poland
| | | | | | - Jacek Olczak
- Molecure S.A., Zwirki i Wigury 101, Warsaw, 02-089, Poland
| | | | | | | |
Collapse
|
10
|
Laubach K, Turan T, Mathew R, Wilsbacher J, Engelhardt J, Samayoa J. Tumor-intrinsic metabolic reprogramming and how it drives resistance to anti-PD-1/PD-L1 treatment. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:611-641. [PMID: 37842241 PMCID: PMC10571065 DOI: 10.20517/cdr.2023.60] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/15/2023] [Accepted: 08/29/2023] [Indexed: 10/17/2023]
Abstract
The development of immune checkpoint blockade (ICB) therapies has been instrumental in advancing the field of immunotherapy. Despite the prominence of these treatments, many patients exhibit primary or acquired resistance, rendering them ineffective. For example, anti-programmed cell death protein 1 (anti-PD-1)/anti-programmed cell death ligand 1 (anti-PD-L1) treatments are widely utilized across a range of cancer indications, but the response rate is only 10%-30%. As such, it is necessary for researchers to identify targets and develop drugs that can be used in combination with existing ICB therapies to overcome resistance. The intersection of cancer, metabolism, and the immune system has gained considerable traction in recent years as a way to comprehensively study the mechanisms that drive oncogenesis, immune evasion, and immunotherapy resistance. As a result, new research is continuously emerging in support of targeting metabolic pathways as an adjuvant to ICB to boost patient response and overcome resistance. Due to the plethora of studies in recent years highlighting this notion, this review will integrate the relevant articles that demonstrate how tumor-derived alterations in energy, amino acid, and lipid metabolism dysregulate anti-tumor immune responses and drive resistance to anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Kyra Laubach
- Computational Oncology, AbbVie, South San Francisco, CA 94080, USA
- Immuno-Oncology, AbbVie, South San Francisco, CA 94080, USA
| | - Tolga Turan
- Computational Oncology, AbbVie, South San Francisco, CA 94080, USA
| | - Rebecca Mathew
- Immuno-Oncology, AbbVie, South San Francisco, CA 94080, USA
| | | | | | - Josue Samayoa
- Computational Oncology, AbbVie, South San Francisco, CA 94080, USA
| |
Collapse
|
11
|
Matulevičiūtė G, Kleizienė N, Račkauskienė G, Martynaitis V, Bieliauskas A, Šachlevičiūtė U, Jankauskas R, Bartkus MR, Sløk FA, Šačkus A. Facile synthesis of new N-(aminocycloalkylene)amino acid compounds using chiral triflate esters with N-Boc-aminopyrrolidines and N-Boc-aminopiperidines. RSC Adv 2023; 13:21378-21394. [PMID: 37469966 PMCID: PMC10353522 DOI: 10.1039/d3ra03060a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023] Open
Abstract
In this study, we prepared a series of new N-(aminocycloalkylene)amino acid derivatives for use in chiral building blocks. The method was based on the conversion of enantiopure α-hydroxy acid esters into the corresponding chiral triflate esters, which were displaced by a nucleophilic substitution SN2 reaction with aminopyrrolidine and aminopiperidine derivatives, and the inversion of the configuration to give methyl 2-[(Boc-amino)cycloamin-1-yl]alkanoates with good yield and high enantiomeric and diastereomeric purity. Synthesized 2-[(Boc-amino)piperidin-1-yl]propanoates combined with ethyl l-phenylalaninate gave new chiral N-Boc- and N-nosyl-dipeptides containing a piperidine moiety. The structures were elucidated by 1H-, 13C-, and 15N-NMR spectroscopy, high-resolution mass spectrometry, and X-ray crystallography analyses.
Collapse
Affiliation(s)
- Gita Matulevičiūtė
- Institute of Synthetic Chemistry, Kaunas University of Technology K. Baršausko g. 59 Kaunas LT-51423 Lithuania
- Department of Organic Chemistry, Kaunas University of Technology Radvilėnų pl. 19 Kaunas LT-50254 Lithuania
| | - Neringa Kleizienė
- Institute of Synthetic Chemistry, Kaunas University of Technology K. Baršausko g. 59 Kaunas LT-51423 Lithuania
| | - Greta Račkauskienė
- Institute of Synthetic Chemistry, Kaunas University of Technology K. Baršausko g. 59 Kaunas LT-51423 Lithuania
| | - Vytas Martynaitis
- Department of Organic Chemistry, Kaunas University of Technology Radvilėnų pl. 19 Kaunas LT-50254 Lithuania
| | - Aurimas Bieliauskas
- Institute of Synthetic Chemistry, Kaunas University of Technology K. Baršausko g. 59 Kaunas LT-51423 Lithuania
| | - Urtė Šachlevičiūtė
- Institute of Synthetic Chemistry, Kaunas University of Technology K. Baršausko g. 59 Kaunas LT-51423 Lithuania
| | - Rokas Jankauskas
- Institute of Synthetic Chemistry, Kaunas University of Technology K. Baršausko g. 59 Kaunas LT-51423 Lithuania
| | - Martynas R Bartkus
- Institute of Synthetic Chemistry, Kaunas University of Technology K. Baršausko g. 59 Kaunas LT-51423 Lithuania
| | - Frank A Sløk
- Vipergen ApS Gammel Kongevej 23A Copenhagen V DK-1610 Denmark
| | - Algirdas Šačkus
- Institute of Synthetic Chemistry, Kaunas University of Technology K. Baršausko g. 59 Kaunas LT-51423 Lithuania
- Department of Organic Chemistry, Kaunas University of Technology Radvilėnų pl. 19 Kaunas LT-50254 Lithuania
| |
Collapse
|
12
|
Park SH, Eun R, Heo J, Lim YT. Nanoengineered drug delivery in cancer immunotherapy for overcoming immunosuppressive tumor microenvironment. Drug Deliv Transl Res 2023; 13:2015-2031. [PMID: 36581707 DOI: 10.1007/s13346-022-01282-8] [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] [Accepted: 12/08/2022] [Indexed: 12/31/2022]
Abstract
Almost like a living being in and of itself, tumors actively interact with and modify their environment to escape immune responses. Owing to the pre-formation of cancer-favorable microenvironment prior to anti-cancer treatment, the numerous attempts that followed propose limited efficacy in oncology. Immunogenicity by activation of immune cells within the tumor microenvironment or recruitment of immune cells from nearby lymph nodes is quickly offset as the immunosuppressive environment, rapidly converting immunogenic cells into immune suppressive cells, overriding the immune system. Tumor cells, as well as regulatory cells, namely M2 macrophages, Treg cells, and MDSCs, derived by the immunosuppressive environment, also cloak from potential anti-tumoral factors by directly or indirectly secreting cytokines, such as IL-10 and TGF-β, related to immune regulation. Enzymes and other metabolic or angiogenetic constituents - VEGF, IDO1, and iNOS - are also employed directed for anti-cancer immune cell malfunctioning. Therefore, the conversion of "cold" immunosuppressive environment into "hot" immune responsive environment is of paramount importance, bestowing the advances in the field of cancer immunotherapy the opportunity to wholly fulfill its intended purpose. This paper reviews the mechanisms by which tumors wield to exercise immune suppression and the nanoengineered delivery strategies being developed to overcome this suppression.
Collapse
Affiliation(s)
- Sei Hyun Park
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Ryounho Eun
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Janghun Heo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea.
| |
Collapse
|
13
|
Jiménez-Alonso JJ, López-Lázaro M. Dietary Manipulation of Amino Acids for Cancer Therapy. Nutrients 2023; 15:2879. [PMID: 37447206 DOI: 10.3390/nu15132879] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Cancer cells cannot proliferate and survive unless they obtain sufficient levels of the 20 proteinogenic amino acids (AAs). Unlike normal cells, cancer cells have genetic and metabolic alterations that may limit their capacity to obtain adequate levels of the 20 AAs in challenging metabolic environments. However, since normal diets provide all AAs at relatively constant levels and ratios, these potentially lethal genetic and metabolic defects are eventually harmless to cancer cells. If we temporarily replace the normal diet of cancer patients with artificial diets in which the levels of specific AAs are manipulated, cancer cells may be unable to proliferate and survive. This article reviews in vivo studies that have evaluated the antitumor activity of diets restricted in or supplemented with the 20 proteinogenic AAs, individually and in combination. It also reviews our recent studies that show that manipulating the levels of several AAs simultaneously can lead to marked survival improvements in mice with metastatic cancers.
Collapse
Affiliation(s)
| | - Miguel López-Lázaro
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Sevilla, Spain
| |
Collapse
|
14
|
Paterek A, Oknińska M, Pilch Z, Sosnowska A, Ramji K, Mackiewicz U, Golab J, Nowis D, Mączewski M. Arginase Inhibition Mitigates Bortezomib-Exacerbated Cardiotoxicity in Multiple Myeloma. Cancers (Basel) 2023; 15:cancers15072191. [PMID: 37046852 PMCID: PMC10093116 DOI: 10.3390/cancers15072191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is associated with increased cardiovascular morbidity and mortality, while MM therapies also result in adverse cardiac effects. Endothelial dysfunction and impaired nitric oxide (NO) pathway is their possible mediator. OBJECTIVE Since MM is associated with increased arginase expression, resulting in the consumption of ʟ-arginine, precursor for NO synthesis, our aim was to test if cardiotoxicity mediated by MM and MM therapeutic, bortezomib (a proteasome inhibitor), can be ameliorated by an arginase inhibitor through improved endothelial function. METHODS We used a mouse Vĸ*MYC model of non-light chain MM. Cardiac function was assessed by echocardiography. RESULTS MM resulted in progressive left ventricular (LV) systolic dysfunction, and bortezomib exacerbated this effect, leading to significant impairment of LV performance. An arginase inhibitor, OAT-1746, protected the heart against bortezomib- or MM-induced toxicity but did not completely prevent the effects of the MM+bortezomib combination. MM was associated with improved endothelial function (assessed as NO production) vs. healthy controls, while bortezomib did not affect it. OAT-1746 improved endothelial function only in healthy mice. NO plasma concentration was increased by OAT-1746 but was not affected by MM or bortezomib. CONCLUSIONS Bortezomib exacerbates MM-mediated LV systolic dysfunction in a mouse model of MM, while an arginase inhibitor partially prevents it. Endothelium does not mediate either these adverse or beneficial effects. This suggests that proteasome inhibitors should be used with caution in patients with advanced myeloma, where the summation of cardiotoxicity could be expected. Therapies aimed at the NO pathway, in particular arginase inhibitors, could offer promise in the prevention/treatment of cardiotoxicity in MM.
Collapse
Affiliation(s)
- Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| | - Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| | - Zofia Pilch
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Anna Sosnowska
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Kavita Ramji
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
- Centre of Preclinical Research, Medical University of Warsaw, 1B Banacha Street, 02-097 Warsaw, Poland
| | - Dominika Nowis
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
- Laboratory of Experimental Medicine, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| |
Collapse
|
15
|
Wu Y, Yang Z, Cheng K, Bi H, Chen J. Small molecule-based immunomodulators for cancer therapy. Acta Pharm Sin B 2022; 12:4287-4308. [PMID: 36562003 PMCID: PMC9764074 DOI: 10.1016/j.apsb.2022.11.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy has led to a paradigm shift in the treatment of cancer. Current cancer immunotherapies are mostly antibody-based, thus possessing advantages in regard to pharmacodynamics (e.g., specificity and efficacy). However, they have limitations in terms of pharmacokinetics including long half-lives, poor tissue/tumor penetration, and little/no oral bioavailability. In addition, therapeutic antibodies are immunogenic, thus may cause unwanted adverse effects. Therefore, researchers have shifted their efforts towards the development of small molecule-based cancer immunotherapy, as small molecules may overcome the above disadvantages associated with antibodies. Further, small molecule-based immunomodulators and therapeutic antibodies are complementary modalities for cancer treatment, and may be combined to elicit synergistic effects. Recent years have witnessed the rapid development of small molecule-based cancer immunotherapy. In this review, we describe the current progress in small molecule-based immunomodulators (inhibitors/agonists/degraders) for cancer therapy, including those targeting PD-1/PD-L1, chemokine receptors, stimulator of interferon genes (STING), Toll-like receptor (TLR), etc. The tumorigenesis mechanism of various targets and their respective modulators that have entered clinical trials are also summarized.
Collapse
Affiliation(s)
| | | | - Kui Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Huichang Bi
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|