1
|
Baird JR, Alice AF, Saito R, Chai Q, Han M, Ng C, Han S, Fernandez B, Ledoux S, Grosse J, Korman AJ, Potuznik M, Rajamanickam V, Bernard B, Crittenden MR, Gough MJ. A novel small molecule Enpp1 inhibitor improves tumor control following radiation therapy by targeting stromal Enpp1 expression. Sci Rep 2024; 14:29913. [PMID: 39622844 PMCID: PMC11612208 DOI: 10.1038/s41598-024-80677-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 11/21/2024] [Indexed: 12/06/2024] Open
Abstract
The uniqueness in each person's cancer cells and variation in immune infiltrates means that each tumor represents a unique problem, but therapeutic targets can be found among their shared features. Radiation therapy alters the interaction between the cancer cells and the stroma through release of innate adjuvants. The extranuclear DNA that can result from radiation damage of cells can result in production of the second messenger cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) by cyclic GMP-AMP synthase (cGAS). In turn, cGAMP can activate the innate sensor stimulator of interferon genes (STING), resulting in innate immune activation. Ectonucleotide pyrophosphatase/phosphodiesterase 1 (Enpp1) is a phosphodiesterase that can be expressed by cancer cells that can degrade cGAMP, thus can decrease or block STING activation following radiation therapy, impairing the innate immunity that is critical to support adaptive immune control of tumors. We observed that many human and murine cancer cells lack Enpp1 expression, but that Enpp1 is expressed in cells of the tumor stroma where it limits tumor control by radiation therapy. We demonstrate in preclinical models the efficacy of a novel Enpp1 inhibitor and show that this inhibitor improves tumor control by radiation even where the cancer cells lack Enpp1. This mechanism requires STING and type I interferon (IFN) receptor expression by non-cancer cells and is dependent on CD8 T cells as a final effector mechanism of tumor control. This suggests that Enpp1 inhibition may be an effective partner for radiation therapy regardless of whether cancer cells express Enpp1. This broadens the potential patient base for whom Enpp1 inhibitors can be applied to improve innate immune responses following radiation therapy.
Collapse
Affiliation(s)
- Jason R Baird
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR, 97213, USA
| | - Alejandro F Alice
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR, 97213, USA
| | - Roland Saito
- VIR Biotechnology Inc, 1800 Owens Street, Suite 900, San Francisco, CA, 94158, USA
| | - Qingqing Chai
- VIR Biotechnology Inc, 1800 Owens Street, Suite 900, San Francisco, CA, 94158, USA
| | - Minhua Han
- VIR Biotechnology Inc, 1800 Owens Street, Suite 900, San Francisco, CA, 94158, USA
| | - Cindy Ng
- VIR Biotechnology Inc, 1800 Owens Street, Suite 900, San Francisco, CA, 94158, USA
| | - Stephanie Han
- VIR Biotechnology Inc, 1800 Owens Street, Suite 900, San Francisco, CA, 94158, USA
| | - Beth Fernandez
- VIR Biotechnology Inc, 1800 Owens Street, Suite 900, San Francisco, CA, 94158, USA
| | - Sarah Ledoux
- VIR Biotechnology Inc, 1800 Owens Street, Suite 900, San Francisco, CA, 94158, USA
| | - Johannes Grosse
- VIR Biotechnology Inc, 1800 Owens Street, Suite 900, San Francisco, CA, 94158, USA
| | - Alan J Korman
- VIR Biotechnology Inc, 1800 Owens Street, Suite 900, San Francisco, CA, 94158, USA
| | - Megan Potuznik
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR, 97213, USA
| | - Venkatesh Rajamanickam
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR, 97213, USA
| | - Brady Bernard
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR, 97213, USA
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR, 97213, USA
- The Oregon Clinic, Portland, OR, 97213, USA
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, Portland, OR, 97213, USA.
| |
Collapse
|
2
|
Crossman BE, Harmon RL, Kostecki KL, McDaniel NK, Iida M, Corday LW, Glitchev CE, Crow MT, Harris MA, Lin CY, Adams JM, Longhurst CA, Nickel KP, Ong IM, Alexandridis RA, Yu M, Yang DT, Hu R, Morris ZS, Hartig GK, Glazer TA, Ramisetty S, Kulkarni P, Salgia R, Kimple RJ, Bruce JY, Harari PM, Wheeler DL. From Bench to Bedside: A Team's Approach to Multidisciplinary Strategies to Combat Therapeutic Resistance in Head and Neck Squamous Cell Carcinoma. J Clin Med 2024; 13:6036. [PMID: 39457986 PMCID: PMC11508784 DOI: 10.3390/jcm13206036] [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: 09/04/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is diagnosed in more than 71,000 patients each year in the United States, with nearly 16,000 associated deaths. One significant hurdle in the treatment of HNSCC is acquired and intrinsic resistance to existing therapeutic agents. Over the past several decades, the University of Wisconsin has formed a multidisciplinary team to move basic scientific discovery along the translational spectrum to impact the lives of HNSCC patients. In this review, we outline key discoveries made throughout the years at the University of Wisconsin to deepen our understanding of therapeutic resistance in HNSCC and how a strong, interdisciplinary team can make significant advances toward improving the lives of these patients by combatting resistance to established therapeutic modalities. We are profoundly grateful to the many scientific teams worldwide whose groundbreaking discoveries, alongside evolving clinical paradigms in head and neck oncology, have been instrumental in making our work possible.
Collapse
Affiliation(s)
- Bridget E. Crossman
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Regan L. Harmon
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Kourtney L. Kostecki
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Nellie K. McDaniel
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Mari Iida
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Luke W. Corday
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Christine E. Glitchev
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Madisen T. Crow
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Madelyn A. Harris
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Candie Y. Lin
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Jillian M. Adams
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Colin A. Longhurst
- Departments of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI 53726, USA; (C.A.L.); (I.M.O.); (R.A.A.)
| | - Kwangok P. Nickel
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Irene M. Ong
- Departments of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI 53726, USA; (C.A.L.); (I.M.O.); (R.A.A.)
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, USA; (R.H.); (J.Y.B.)
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53705, USA
| | - Roxana A. Alexandridis
- Departments of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI 53726, USA; (C.A.L.); (I.M.O.); (R.A.A.)
| | - Menggang Yu
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA;
| | - David T. Yang
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA;
| | - Rong Hu
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, USA; (R.H.); (J.Y.B.)
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA;
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
| | - Gregory K. Hartig
- Department of Surgery, University of Wisconsin, Madison, WI 53705, USA; (G.K.H.); (T.A.G.)
| | - Tiffany A. Glazer
- Department of Surgery, University of Wisconsin, Madison, WI 53705, USA; (G.K.H.); (T.A.G.)
| | - Sravani Ramisetty
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (S.R.); (P.K.); (R.S.)
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (S.R.); (P.K.); (R.S.)
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (S.R.); (P.K.); (R.S.)
| | - Randall J. Kimple
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, USA; (R.H.); (J.Y.B.)
| | - Justine Y. Bruce
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, USA; (R.H.); (J.Y.B.)
- Department of Medicine, University of Wisconsin, Madison, WI 53705, USA
| | - Paul M. Harari
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, USA; (R.H.); (J.Y.B.)
| | - Deric L. Wheeler
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705, USA; (B.E.C.); (R.L.H.); (M.I.); (M.A.H.); (C.Y.L.); (K.P.N.); (Z.S.M.); (R.J.K.); (P.M.H.)
- Carbone Cancer Center, University of Wisconsin, Madison, WI 53792, USA; (R.H.); (J.Y.B.)
| |
Collapse
|
3
|
Frey RR, Jana N, Gorman JV, Wang J, Smith HA, Bromberg KD, Thakur A, Doktor SZ, Indulkar AS, Jakob CG, Upadhyay AK, Qiu W, Manaves V, Gambino F, Valentino SA, Montgomery D, Zhou Y, Li T, Buchanan FG, Ferguson DC, Kurnick MD, Kapecki N, Lai A, Michaelides MR, Penning TD. Discovery of Potent Azetidine-Benzoxazole MerTK Inhibitors with In Vivo Target Engagement. J Med Chem 2024; 67:17033-17052. [PMID: 39350472 DOI: 10.1021/acs.jmedchem.4c01451] [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: 10/11/2024]
Abstract
Inhibition of the receptor tyrosine kinase MerTK by small molecules has the potential to augment the immune response to tumors. Potent, selective inhibitors with high levels of in vivo target engagement are needed to fully evaluate the potential use of MerTK inhibitors as cancer therapeutics. We report the discovery and optimization of a series of pyrazinamide-based type 1.5 MerTK inhibitors bearing an azetidine-benzoxazole substituent. Compound 31 potently engages the target in vivo and demonstrates single agent activity in the immune-driven MC-38 murine syngeneic tumor model.
Collapse
Affiliation(s)
- Robin R Frey
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Navendu Jana
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Jacob V Gorman
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Jin Wang
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Heath A Smith
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Kenneth D Bromberg
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Ashish Thakur
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Stella Z Doktor
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Anura S Indulkar
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Clarissa G Jakob
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Anup K Upadhyay
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Wei Qiu
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Vlasios Manaves
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Frank Gambino
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Stephen A Valentino
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Debra Montgomery
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Yebin Zhou
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Tao Li
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Fritz G Buchanan
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Debra C Ferguson
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Matthew D Kurnick
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Nicolas Kapecki
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Albert Lai
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Michael R Michaelides
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Thomas D Penning
- Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| |
Collapse
|
4
|
Vázquez-Bellón N, Martínez-Bosch N, García de Frutos P, Navarro P. Hallmarks of pancreatic cancer: spotlight on TAM receptors. EBioMedicine 2024; 107:105278. [PMID: 39137571 PMCID: PMC11367522 DOI: 10.1016/j.ebiom.2024.105278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/14/2024] [Accepted: 07/29/2024] [Indexed: 08/15/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents the most prevalent type of pancreatic cancer and ranks among the most aggressive tumours, with a 5-year survival rate of less than 11%. Projections indicate that by 2030, it will become the second leading cause of cancer-related deaths. PDAC presents distinctive hallmarks contributing to its dismal prognosis: (i) late diagnosis, (ii) heterogenous and complex mutational landscape, (iii) high metastatic potential, (iv) dense fibrotic stroma, (v) immunosuppressive microenvironment, and (vi) high resistance to therapy. Mounting evidence has shown a role for TAM (Tyro3, AXL, MerTK) family of tyrosine kinase receptors in PDAC initiation and progression. This review aims to describe the impact of TAM receptors on the defining hallmarks of PDAC and discuss potential future directions using these proteins as novel biomarkers for early diagnosis and targets for precision therapy in PDAC, an urgent unmet clinical need.
Collapse
Affiliation(s)
- Núria Vázquez-Bellón
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB)-CSIC and Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain; PhD Program in Biomedicine, Facultat de Medicina (Campus Clínic), Universitat de Barcelona, Barcelona, Spain
| | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Research Institute (HMRI), Unidad Asociada IIBB-CSIC, Barcelona, Spain
| | - Pablo García de Frutos
- Department of Cell Death and Proliferation, IIBB-CSIC, Unidad Asociada IMIM/IIBB-CSIC, Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), and IDIBAPS, Barcelona, Spain.
| | - Pilar Navarro
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB)-CSIC and Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain; Cancer Research Program, Hospital del Mar Research Institute (HMRI), Unidad Asociada IIBB-CSIC, Barcelona, Spain.
| |
Collapse
|
5
|
Ghosh S, Finnemann SC, Vollrath D, Rothlin CV. In the Eyes of the Beholder-New Mertk Knockout Mouse and Re-Evaluation of Phagocytosis versus Anti-Inflammatory Functions of MERTK. Int J Mol Sci 2024; 25:5299. [PMID: 38791338 PMCID: PMC11121519 DOI: 10.3390/ijms25105299] [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: 03/16/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Greg Lemke's laboratory was one of the pioneers of research into the TAM family of receptor tyrosine kinases (RTKs). Not only was Tyro3 cloned in his laboratory, but his group also extensively studied mice knocked out for individual or various combinations of the TAM RTKs Tyro3, Axl, and Mertk. Here we primarily focus on one of the paralogs-MERTK. We provide a historical perspective on rodent models of loss of Mertk function and their association with retinal degeneration and blindness. We describe later studies employing mouse genetics and the generation of newer knockout models that point out incongruencies with the inference that loss of MERTK-dependent phagocytosis is sufficient for severe, early-onset photoreceptor degeneration in mice. This discussion is meant to raise awareness with regards to the limitations of the original Mertk knockout mouse model generated using 129 derived embryonic stem cells and carrying 129 derived alleles and the role of these alleles in modifying Mertk knockout phenotypes or even displaying Mertk-independent phenotypes. We also suggest molecular approaches that can further Greg Lemke's scintillating legacy of dissecting the molecular functions of MERTK-a protein that has been described to function in phagocytosis as well as in the negative regulation of inflammation.
Collapse
Affiliation(s)
- Sourav Ghosh
- Department of Neurology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Silvia C. Finnemann
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA;
| | - Douglas Vollrath
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Carla V. Rothlin
- Department of Pharmacology, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
6
|
Lorimer IAJ. Potential roles for efferocytosis in glioblastoma immune evasion. Neurooncol Adv 2024; 6:vdae012. [PMID: 38616895 PMCID: PMC11012614 DOI: 10.1093/noajnl/vdae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024] Open
Abstract
Glioblastoma is an aggressive and incurable brain cancer. This cancer establishes both local and systemic immunosuppression that creates a major obstacle to effective immunotherapies. Many studies point to tumor-resident myeloid cells (primarily microglia and macrophages) as key mediators of this immunosuppression. Myeloid cells exhibit a high level of plasticity with respect to their phenotype and are capable of both stimulating and repressing immune responses. How glioblastomas recruit myeloid cells and exploit them to avoid the immune system is an active area of research. Macrophages can acquire an immunosuppressive phenotype as a consequence of exposure to cytokines such as TGFB1 or IL4; in addition, macrophages can acquire an immunosuppressive phenotype as a consequence of the engulfment of apoptotic cells, a process referred to as efferocytosis. There is substantial evidence that glioblastoma cells are able to secrete cytokines and other factors that induce an immunosuppressive phenotype in macrophages and microglia. However, less is known about the contribution of efferocytosis to immunosuppression in glioblastoma. Here I review the literature in this area and discuss the potential of efferocytosis inhibition to improve glioblastoma response to immunotherapy.
Collapse
Affiliation(s)
- Ian A J Lorimer
- Cancer Research Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
7
|
Qiu H, Shao Z, Wen X, Liu Z, Chen Z, Qu D, Ding X, Zhang L. Efferocytosis: An accomplice of cancer immune escape. Biomed Pharmacother 2023; 167:115540. [PMID: 37741255 DOI: 10.1016/j.biopha.2023.115540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/13/2023] [Accepted: 09/17/2023] [Indexed: 09/25/2023] Open
Abstract
The clearance of apoptotic cells by efferocytes such as macrophages and dendritic cells is termed as "efferocytosis", it plays critical roles in maintaining tissue homeostasis in multicellular organisms. Currently, available studies indicate that efferocytosis-related molecules and pathways are tightly associated with cancer development, metastasis and treatment resistance, efferocytosis also induces an immunosuppressive tumor microenvironment and assists cancer cells escape from immune surveillance. In this study, we reviewed the underlying mechanisms of efferocytosis in mediating the occurrence of cancer immune escape, and then emphatically summarized the strategies of using efferocytosis as therapeutic target to enhance the anti-tumor efficacies of immune checkpoint inhibitors, hoping to provide powerful evidences for more effective therapeutic regimens of malignant tumors.
Collapse
Affiliation(s)
- Hui Qiu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhiying Shao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin Wen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhengyang Liu
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ziqin Chen
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Debao Qu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xin Ding
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Longzhen Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
8
|
Chen JLY, Yang SJ, Pan CK, Lin LC, Tsai CY, Wang CH, Huang YS, Lin YL, Kuo SH, Shieh MJ. Cisplatin and Albumin-Based Gold-Cisplatin Nanoparticles Enhance Ablative Radiation Therapy-Induced Antitumor Immunity in Local and Distant Tumor Microenvironment. Int J Radiat Oncol Biol Phys 2023; 116:1135-1149. [PMID: 36792014 DOI: 10.1016/j.ijrobp.2023.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 02/15/2023]
Abstract
PURPOSE Ablative radiation therapy (RT) is an important strategy to eliminate primary tumor and can potentially induce the abscopal effect. Human serum albumin nanoparticle (NP) was used for controlled release of cisplatin to decrease cisplatin's systemic toxicity, and gold (Au) was added to increase RT-induced immunogenic cell death and potentiate the abscopal antitumor immunity. METHODS AND MATERIALS The designed albumin-based cisplatin-conjugated AuNPs were administered concurrently with ablative RT. C57BL/6 mice implanted with syngeneic murine Lewis lung carcinoma or murine MB49 tumor models were treated with ablative RT (12 Gy per fraction for 2 fractions, total 24 Gy), cisplatin, or Au-cisplatin NPs. RESULTS Combining ablative RT with cisplatin or Au-cisplatin NPs both destroyed the primary tumor effectively and elicited immunogenic cell death accompanied by release of danger-associated molecular patterns. This enhanced recruitment of effector tumor-infiltrating immune cells, including natural killer T cells and CD8+ T cells, and elicited an increased percentage of professional antigen-presenting CD11c+ dendritic cells. Transient weight loss, accompanying hepatotoxicity, nephrotoxicity, and hematopoietic suppression, was observed as a systemic adverse event in the cisplatin but not the Au-cisplatin NPs group. Cisplatin and Au-cisplatin NPs both showed equivalent ability to reduce metastatic potential when combined with ablative RT, confirmed by suppressed unirradiated flank tumor growth and decreased metastatic lung tumor burden, which translated to improved survival. Mobilization and abundance of effector tumor-infiltrating immune cells including CD8+ T cells and dendritic cells were observed in the distant lung tumor microenvironment after ablative RT with cisplatin or Au-cisplatin NPs, demonstrating increased antitumor immunotherapeutic activity as an abscopal effect. CONCLUSIONS Compared with cisplatin, the albumin-based Au-cisplatin NPs exhibited equivalent but no superior antitumor immunotherapeutic activity while reducing systemic adverse events and can be safely administered concurrently with ablative RT. Alternative NP formulations may be designed to further improve anticancer outcomes.
Collapse
Affiliation(s)
- Jenny Ling-Yu Chen
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan; Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan; Department of Radiation Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Shu-Jyuan Yang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| | - Chun-Kai Pan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Cheng Lin
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Yi Tsai
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan; Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | - Yu-Sen Huang
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Li Lin
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan.
| | - Sung-Hsin Kuo
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan; Department of Radiation Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Ming-Jium Shieh
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
9
|
Sharon S, Daher-Ghanem N, Zaid D, Gough MJ, Kravchenko-Balasha N. The immunogenic radiation and new players in immunotherapy and targeted therapy for head and neck cancer. FRONTIERS IN ORAL HEALTH 2023; 4:1180869. [PMID: 37496754 PMCID: PMC10366623 DOI: 10.3389/froh.2023.1180869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023] Open
Abstract
Although treatment modalities for head and neck cancer have evolved considerably over the past decades, survival rates have plateaued. The treatment options remained limited to definitive surgery, surgery followed by fractionated radiotherapy with optional chemotherapy, and a definitive combination of fractionated radiotherapy and chemotherapy. Lately, immunotherapy has been introduced as the fourth modality of treatment, mainly administered as a single checkpoint inhibitor for recurrent or metastatic disease. While other regimens and combinations of immunotherapy and targeted therapy are being tested in clinical trials, adapting the appropriate regimens to patients and predicting their outcomes have yet to reach the clinical setting. Radiotherapy is mainly regarded as a means to target cancer cells while minimizing the unwanted peripheral effect. Radiotherapy regimens and fractionation are designed to serve this purpose, while the systemic effect of radiation on the immune response is rarely considered a factor while designing treatment. To bridge this gap, this review will highlight the effect of radiotherapy on the tumor microenvironment locally, and the immune response systemically. We will review the methodology to identify potential targets for therapy in the tumor microenvironment and the scientific basis for combining targeted therapy and radiotherapy. We will describe a current experience in preclinical models to test these combinations and propose how challenges in this realm may be faced. We will review new players in targeted therapy and their utilization to drive immunogenic response against head and neck cancer. We will outline the factors contributing to head and neck cancer heterogeneity and their effect on the response to radiotherapy. We will review in-silico methods to decipher intertumoral and intratumoral heterogeneity and how these algorithms can predict treatment outcomes. We propose that (a) the sequence of surgery, radiotherapy, chemotherapy, and targeted therapy should be designed not only to annul cancer directly, but to prime the immune response. (b) Fractionation of radiotherapy and the extent of the irradiated field should facilitate systemic immunity to develop. (c) New players in targeted therapy should be evaluated in translational studies toward clinical trials. (d) Head and neck cancer treatment should be personalized according to patients and tumor-specific factors.
Collapse
Affiliation(s)
- Shay Sharon
- Department of Oral and Maxillofacial Surgery, Hadassah Medical Center, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Oral and Maxillofacial Surgery, Boston University and Boston Medical Center, Boston, MA, United States
| | - Narmeen Daher-Ghanem
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Deema Zaid
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
10
|
Redmond WL. Challenges and opportunities in the development of combination immunotherapy with OX40 agonists. Expert Opin Biol Ther 2023; 23:901-912. [PMID: 37587644 PMCID: PMC10530613 DOI: 10.1080/14712598.2023.2249396] [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: 07/18/2023] [Accepted: 08/15/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION Costimulatory members of the tumor necrosis factor receptor family, such as OX40 (CD134), provide essential survival and differentiation signals that enhance T cell function. Specifically, OX40 (CD134) agonists stimulate potent anti-tumor immunity in a variety of preclinical models but their therapeutic impact in patients with advanced malignancies has been limited thus far. AREAS COVERED In this review, we discuss the current state of combination immunotherapy with OX40 agonists including preclinical studies and recent clinical trials. We also discuss the strengths and limitations of these approaches and provide insight into alternatives that may help enhance the efficacy of combination OX40 agonist immunotherapy. EXPERT OPINION OX40 agonist immunotherapy has not yet demonstrated significant clinical activity as a monotherapy or in combination with immune checkpoint blockade (ICB), likely due to several factors including the timing of administration, drug potency, and selection of agents for combination therapy clinical trials. We believe that careful consideration of the biological mechanisms regulating OX40 expression and function may help inform new approaches, particularly in combination with novel agents, capable of increasing the therapeutic efficacy of this approach.
Collapse
Affiliation(s)
- William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, 4805 NE Glisan St., 2N35, Portland, OR, 97213
| |
Collapse
|
11
|
Huang H, Jiang J, Chen R, Lin Y, Chen H, Ling Q. The role of macrophage TAM receptor family in the acute-to-chronic progression of liver disease: From friend to foe? Liver Int 2022; 42:2620-2631. [PMID: 35900248 DOI: 10.1111/liv.15380] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/12/2022]
Abstract
Hepatic macrophages, the key cellular components of the liver, emerge as essential players in liver inflammation, tissue repair and subsequent fibrosis, as well as tumorigenesis. Recently, the TAM receptor tyrosine kinase family, consisting of Tyro3, Axl and MerTK, was found to be a pivotal modulator of macrophages. Activation of macrophage TAM receptor signalling promotes the efferocytosis of apoptotic cells and skews the polarization of macrophages. After briefly reviewing the mechanisms of TAM receptor signalling in macrophage polarization, we focus on their role in liver diseases from acute injury to chronic inflammation, fibrosis and then to tumorigenesis. Notably, macrophage TAM receptor signalling seems to be a two-edged sword for liver diseases. On one hand, the activation of TAM receptor signalling inhibits inflammation and facilitates tissue repair during acute liver injury. On the other hand, continuous activation of the signalling contributes to the process of chronic inflammation into fibrosis and tumorigenesis by evoking hepatic stellate cells and inhibiting anti-tumour immunity. Therefore, targeting macrophage TAM receptors and clarifying its downstream pathways will be exciting prospects for the precaution and treatment of liver diseases, particularly at different stages or statuses.
Collapse
Affiliation(s)
- Haitao Huang
- Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Jingyu Jiang
- Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Ruihan Chen
- Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Yimou Lin
- Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Hui Chen
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Qi Ling
- Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China
| |
Collapse
|
12
|
Capistrano I R, Paul S, Boere I, Pantziarka P, Chopra S, Nout RA, Bouche G. Drug repurposing as a potential source of innovative therapies in cervical cancer. Int J Gynecol Cancer 2022; 32:1377-1386. [PMID: 36137575 PMCID: PMC9664115 DOI: 10.1136/ijgc-2022-003585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/19/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Drug repurposing is an alternative development pathway that utilizes the properties of drugs approved for other diseases and builds on available safety and pharmacological data to develop the drug as a potential treatment for other diseases. A literature-based approach was performed to identify drug repurposing opportunities in cervical cancer to inform future research and trials. METHODS We queried PubMed for each drug included in two databases (ReDO_DB and CDcervix_DB, which include 300+ non-cancer drugs and 200+ cancer drugs not used in cervical cancer, respectively) and manually assessed all abstracts for relevance and activity in cervix cancer, and type of evidence. Subsequently, we also performed a search of clinical trial databases where we generated a list of registered trials in cervical cancer with all drugs from our databases. RESULTS Of the 534 drugs from both databases, 174 (33%) had at least one relevant abstract or registered trial in cervical cancer. 94 (18%) drugs had at least human data available, and 52 (10%) drugs were evaluated in registered trials. To prioritize drugs to consider for future trials, all 174 drugs were further assessed for strength of scientific rationale, feasibility for integration in cervical cancer standard of care, evidence of radiosensitization, and potential mechanism of action. Out of the 174 drugs, 38 (22%) potential drug candidates were selected. CONCLUSION This study resulted in a list of candidate drugs for potential evaluation in cervical cancer. Many drugs might warrant additional (pre)clinical investigation, which could be done in a coordinated manner using platform trials.
Collapse
Affiliation(s)
| | - Sonz Paul
- Department of Radiation Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Ingrid Boere
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Pan Pantziarka
- Anticancer Fund, Meise, Belgium
- The George Pantziarka TP53 Trust, London, UK
| | - Supriya Chopra
- Department of Radiation Oncology, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Navi Mumbai, India
| | - Remi A Nout
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | | |
Collapse
|
13
|
Blair T, Baird J, Bambina S, Kramer G, Gostissa M, Harvey CJ, Gough MJ, Crittenden MR. ICOS is upregulated on T cells following radiation and agonism combined with radiation results in enhanced tumor control. Sci Rep 2022; 12:14954. [PMID: 36056093 PMCID: PMC9440216 DOI: 10.1038/s41598-022-19256-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/26/2022] [Indexed: 01/21/2023] Open
Abstract
Multiple preclinical studies have shown improved outcomes when radiation therapy is combined with immune modulating antibodies. However, to date, many of these promising results have failed to translate to successful clinical studies. This led us to explore additional checkpoint and co-stimulatory pathways that may be regulated by radiation therapy. Here, we demonstrate that radiation increases the expression of inducible T cell co-stimulator (ICOS) on both CD4 and CD8 T cells in the blood following treatment. Moreover, when we combined a novel ICOS agonist antibody with radiation we observed durable cures across multiple tumor models and mouse strains. Depletion studies revealed that CD8 T cells were ultimately required for treatment efficacy, but CD4 T cells and NK cells also partially contributed to tumor control. Phenotypic analysis showed that the combination therapy diminished the increased infiltration of regulatory T cells into the tumor that typically occurs following radiation alone. Finally, we demonstrate in a poorly immunogenic pancreatic tumor model which is resistant to combined radiation and anti-PD1 checkpoint blockade that the addition of this novel ICOS agonist antibody to the treatment regimen results in tumor control. These findings identify ICOS as part of a T cell pathway that is modulated by radiation and targeting this pathway with a novel ICOS antibody results in durable tumor control in preclinical models.
Collapse
Affiliation(s)
- Tiffany Blair
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, North Pavilion, Suite 2N108, Portland, OR, 97213, USA
| | - Jason Baird
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, North Pavilion, Suite 2N108, Portland, OR, 97213, USA
| | - Shelly Bambina
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, North Pavilion, Suite 2N108, Portland, OR, 97213, USA
| | - Gwen Kramer
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, North Pavilion, Suite 2N108, Portland, OR, 97213, USA
| | - Monica Gostissa
- Jounce Therapeutics, Inc., 780 Memorial Drive, Cambridge, MA, 02139, USA
| | - Christopher J Harvey
- Jounce Therapeutics, Inc., 780 Memorial Drive, Cambridge, MA, 02139, USA
- Phenomic AI, 661 University Ave Suite 1300, Toronto, ON, M5G 0B7, Canada
| | - Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, North Pavilion, Suite 2N108, Portland, OR, 97213, USA
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St, North Pavilion, Suite 2N108, Portland, OR, 97213, USA.
- The Oregon Clinic, Portland, OR, 97213, USA.
| |
Collapse
|
14
|
Gough MJ, Crittenden MR. The paradox of radiation and T cells in tumors. Neoplasia 2022; 31:100808. [PMID: 35691060 PMCID: PMC9194456 DOI: 10.1016/j.neo.2022.100808] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/05/2022] [Accepted: 05/13/2022] [Indexed: 10/27/2022]
Abstract
In this review we consider what appears to be a paradox in immunotherapies based around radiation therapy. The paradox is based on three main points. 1. That T cells are needed for radiation's efficacy; 2. That tumor-specific T cells are enriched in the field of treatment; and 3. That radiation kills T cells in the treatment field. We discuss evidence of the effect of radiation on T cells in the field given their ongoing movement in and out of tissues and the tumor, and how the movement of T cells impacts the treated primary tumor and untreated distant metastases. Given this evidence, we revisit the paradox to understand how the extraordinary efficacy of radiation and immunity in preclinical models is dependent on this radiation sensitive cell.
Collapse
Affiliation(s)
- Michael J Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St., Portland, OR 97213, USA.
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, 4805 NE Glisan St., Portland, OR 97213, USA; The Oregon Clinic, Portland, OR, 97213, USA
| |
Collapse
|
15
|
Akalu YT, Mercau ME, Ansems M, Hughes LD, Nevin J, Alberto EJ, Liu XN, He LZ, Alvarado D, Keler T, Kong Y, Philbrick WM, Bosenberg M, Finnemann SC, Iavarone A, Lasorella A, Rothlin CV, Ghosh S. Tissue-specific modifier alleles determine Mertk loss-of-function traits. eLife 2022; 11:80530. [PMID: 35969037 PMCID: PMC9433089 DOI: 10.7554/elife.80530] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/13/2022] [Indexed: 11/19/2022] Open
Abstract
Knockout (KO) mouse models play critical roles in elucidating biological processes behind disease-associated or disease-resistant traits. As a presumed consequence of gene KO, mice display certain phenotypes. Based on insight into the molecular role of said gene in a biological process, it is inferred that the particular biological process causally underlies the trait. This approach has been crucial towards understanding the basis of pathological and/or advantageous traits associated with Mertk KO mice. Mertk KO mice suffer from severe, early-onset retinal degeneration. MERTK, expressed in retinal pigment epithelia, is a receptor tyrosine kinase with a critical role in phagocytosis of apoptotic cells or cellular debris. Therefore, early-onset, severe retinal degeneration was described to be a direct consequence of failed MERTK-mediated phagocytosis of photoreceptor outer segments by retinal pigment epithelia. Here, we report that the loss of Mertk alone is not sufficient for retinal degeneration. The widely used Mertk KO mouse carries multiple coincidental changes in its genome that affect the expression of a number of genes, including the Mertk paralog Tyro3. Retinal degeneration manifests only when the function of Tyro3 is concomitantly lost. Furthermore, Mertk KO mice display improved anti-tumor immunity. MERTK is expressed in macrophages. Therefore, enhanced anti-tumor immunity was inferred to result from the failure of macrophages to dispose of cancer cell corpses, resulting in a pro-inflammatory tumor microenvironment. The resistance against two syngeneic mouse tumor models observed in Mertk KO mice is not, however, phenocopied by the loss of Mertk alone. Neither Tyro3 nor macrophage phagocytosis by alternate genetic redundancy accounts for the absence of anti-tumor immunity. Collectively, our results indicate that context-dependent epistasis of independent modifier alleles determines Mertk KO traits.
Collapse
Affiliation(s)
- Yemsratch T Akalu
- Department of Immunobiology, Yale School of MedicineNew HavenUnited States
| | - Maria E Mercau
- Department of Immunobiology, Yale School of MedicineNew HavenUnited States
| | - Marleen Ansems
- Department of Immunobiology, Yale School of MedicineNew HavenUnited States
| | - Lindsey D Hughes
- Department of Immunobiology, Yale School of MedicineNew HavenUnited States
| | - James Nevin
- Department of Immunobiology, Yale School of MedicineNew HavenUnited States
| | - Emily J Alberto
- Department of Immunobiology, Yale School of MedicineNew HavenUnited States
| | - Xinran N Liu
- Department of Cell Biology, Center for Cellular and Molecular Imaging, Yale School of MedicineNew HavenUnited States
| | - Li-Zhen He
- Celldex TherapeuticsNew HavenUnited States
| | | | | | - Yong Kong
- Department of Molecular Biophysics and Biochemistry, W. M. Keck Foundation Biotechnology Resource Laboratory, School of Medicine, Yale UniversityNew HavenUnited States
| | - William M Philbrick
- Center on Endocrinology and Metabolism, Yale Genome Editing Center, School of Medicine, Yale UniversityNew HavenUnited States
| | - Marcus Bosenberg
- Departments of Dermatology, Pathology and Immunobiology, Yale School of MedicineNew HavenUnited States
| | - Silvia C Finnemann
- Center for Cancer, Genetic Diseases and Gene Regulation, Department of Biological Sciences, Fordham UniversityBronxUnited States
| | - Antonio Iavarone
- Departments of Neurology and Pathology and Cell Biology, Institute for Cancer Genetics, Columbia Medical CenterNew YorkUnited States
| | - Anna Lasorella
- Departments of Pediatrics and Pathology and Cell Biology, Institute for Cancer Genetics, Columbia UniversityNew YorkUnited States
| | - Carla V Rothlin
- Departments of Immunobiology and Pharmacology, Yale School of MedicineNew HavenUnited States
| | - Sourav Ghosh
- Departments of Neurology and Pharmacology, Yale School of MedicineNew HavenUnited States
| |
Collapse
|
16
|
Two-Front War on Cancer-Targeting TAM Receptors in Solid Tumour Therapy. Cancers (Basel) 2022; 14:cancers14102488. [PMID: 35626092 PMCID: PMC9140196 DOI: 10.3390/cancers14102488] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary In recent years, many studies have shown the importance of TAM kinases in both normal and neoplastic cells. In this review, we present and discuss the role of the TAM family (AXL, MERTK, TYRO3) of receptor tyrosine kinases (RTKs) as a dual target in cancer, due to their intrinsic roles in tumour cell survival, migration, chemoresistance, and their immunosuppressive roles in the tumour microenvironment. This review presents the potential of TAMs as emerging therapeutic targets in cancer treatment, focusing on the distinct structures of TAM receptor tyrosine kinases. We analyse and compare different strategies of TAM inhibition, for a full perspective of current and future battlefields in the war with cancer. Abstract Receptor tyrosine kinases (RTKs) are transmembrane receptors that bind growth factors and cytokines and contain a regulated kinase activity within their cytoplasmic domain. RTKs play an important role in signal transduction in both normal and malignant cells, and their encoding genes belong to the most frequently affected genes in cancer cells. The TAM family proteins (TYRO3, AXL, and MERTK) are involved in diverse biological processes: immune regulation, clearance of apoptotic cells, platelet aggregation, cell proliferation, survival, and migration. Recent studies show that TAMs share overlapping functions in tumorigenesis and suppression of antitumour immunity. MERTK and AXL operate in innate immune cells to suppress inflammatory responses and promote an immunosuppressive tumour microenvironment, while AXL expression correlates with epithelial-to-mesenchymal transition, metastasis, and motility in tumours. Therefore, TAM RTKs represent a dual target in cancer due to their intrinsic roles in tumour cell survival, migration, chemoresistance, and their immunosuppressive roles in the tumour microenvironment (TME). In this review, we discuss the potential of TAMs as emerging therapeutic targets in cancer treatment. We critically assess and compare current approaches to target TAM RTKs in solid tumours and the development of new inhibitors for both extra- and intracellular domains of TAM receptor kinases.
Collapse
|
17
|
Zhang J, Dai Z, Yan C, Wang D, Tang D. Blocking antibody-mediated phosphatidylserine enhances cancer immunotherapy. J Cancer Res Clin Oncol 2021; 147:3639-3651. [PMID: 34499223 DOI: 10.1007/s00432-021-03792-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy is a major breakthrough in tumor therapy and has been used in monotherapy or combination therapy. However, it has been associated with poor immune tolerance in some patients or immune-related adverse events. Therefore, ideal and reliable tumor elimination strategies are urgently needed to overcome these shortcomings. Phosphatidylserine (PS) is a negatively charged phospholipid, usually present in the inner lobules of eukaryotic cell membranes. Under certain physiological or pathological conditions, PS may be exposed on the outer leaflets of apoptotic cells serving as recognition signals by phagocytes and modulating the immune response. On the contrary, increased exposure of PS in the tumor microenvironment can significantly antagonize the body's anti-tumor immunity, thereby promoting tumor growth and metastasis. During radiotherapy and chemotherapy, PS-mediated immunosuppression increases the PS levels in necrotic tissue in the tumor microenvironment, further suppressing tumor immunity. PS-targeted therapy is a promising strategy in cancer immunotherapy. It inhibits tumor growth and improves the anti-tumor activity of immune checkpoint inhibitors. A comprehensive understanding of the mechanism of PS-targeted therapy opens up a new perspective for future cancer immunotherapies.
Collapse
Affiliation(s)
- Jie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Zhujiang Dai
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Cheng Yan
- Dalian Medical University, Dalian, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225001, People's Republic of China.
| |
Collapse
|
18
|
Xiao Y, Zhang L, Zhu J, Zhang Y, Yang R, Yan J, Huang R, Zheng C, Xiao W, Huang C, Wang Y. Predicting the herbal medicine triggering innate anti-tumor immunity from a system pharmacology perspective. Biomed Pharmacother 2021; 143:112105. [PMID: 34560533 DOI: 10.1016/j.biopha.2021.112105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 01/22/2023] Open
Abstract
Although the main focus of immuno-oncology has been manipulating the adaptive immune system, tumor associated macrophages (TAMs) are the main infiltrating component in the tumor microenvironment (TME) and play a critical role in cancer progression. TAMs are mainly divided into two different subtypes: macrophages with antitumor or killing activity are called M1 while tumor-promoting or healing macrophages are named M2. Therefore, controlling the polarization of TAMs is an important strategy for cancer treatment, but there is no particularly effective means to regulate the polarization process. Here, combined systems pharmacology targets and pathways analysis strategy, we uncovered Scutellariae Radix (SR) has the potential to regulate TAMs polarization to inhibit the growth of non-small cell lung cancer (NSCLC). Firstly, systems pharmacology approach was used to reveal the active components of SR targeting macrophages in TME through compound target prediction and target-microenvironment phenotypic association analysis. Secondly, in vitro experiment verified that WBB (wogonin, baicalein and baicalin), major active ingredients of SR are significantly related to macrophages and survival, initiated macrophages programming to M1-like macrophages to promoted the apoptosis of tumor cells. Finally, we evidenced that WBB effectively inhibited tumor growth in LLC (Lewis lung carcinoma) tumor-bearing mice and increased the infiltration of M1-type macrophages in TME. Overall, the systems pharmacology strategy offers a paradigm to understand the mechanism of polypharmacology of natural products targeting TME.
Collapse
Affiliation(s)
- Yue Xiao
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| | - Lulu Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| | - Jinglin Zhu
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| | - Yuru Zhang
- Pharmacology Department, School of Pharmacy, Shihezi University, Shihezi, China.
| | - Ruijie Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| | - Jiangna Yan
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| | - Ruifei Huang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| | - Chunli Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China.
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Parmaceutical Co. Ltd., Lianyungang, China.
| | - Chao Huang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China; Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, China.
| | - Yonghua Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China; Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, China.
| |
Collapse
|
19
|
Yin X, Luo J, Xu C, Meng C, Zhang J, Yu H, Liu N, Yuan Z, Wang P, Sun Y, Zhao L. Is a higher estimated dose of radiation to immune cells predictive of survival in patients with locally advanced non-small cell lung cancer treated with thoracic radiotherapy? Radiother Oncol 2021; 159:218-223. [PMID: 33798612 DOI: 10.1016/j.radonc.2021.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/14/2021] [Accepted: 03/18/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE In previous studies, the estimated dose of radiation to immune cells (EDRIC) showed a correlation with overall survival (OS) of patients with locally advanced non-small cell lung cancer (LA-NSCLC) who received thoracic radiotherapy. However, several factors such as gross tumor volume (GTV) and lymph node (N) stage may impact EDRIC. The purpose of this study was to identify the factors influencing EDRIC and to further assess the prognostic relevance of EDRIC. MATERIALS AND METHODS We retrospectively analyzed 201 patients with LA-NSCLC who received radiotherapy between 2012 and 2017. EDRIC was calculated based on the model developed by Jin et al. Kaplan-Meier method and Cox proportional hazards regression were used to analyze the correlation of potential factors with OS, local progression-free survival (LPFS), and distant metastasis-free survival (DMFS). Spearman's rank correlation was used to assess the correlation between variables. RESULTS Both GTV and N stage showed a positive correlation with EDRIC (r = 0.347, P < 0.001 and r = 0.249, P < 0.001, respectively). EDRIC was independently associated with DMFS (HR 1.185, P < 0.001). GTV was associated with OS (HR 1.006, P < 0.001), LPFS (HR 1.003, P = 0.017), and DMFS (HR 1.003, P = 0.032). While using GTV as a stratification factor in Kaplan-Meier analysis, EDRIC showed a trend of negative correlation with OS in GTV ≤ 66.6 cm3 group (P = 0.061). CONCLUSION EDRIC was an independent prognostic factor for metastasis and it was affected by GTV and N stage. However, the effect of EDRIC on OS was influenced by GTV.
Collapse
Affiliation(s)
- Xiaoming Yin
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, China; Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, China
| | - Jing Luo
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, China
| | - Cai Xu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, China
| | - Chunliu Meng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, China
| | - Jiaqi Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, China
| | - Hao Yu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, China
| | - Ningbo Liu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, China
| | - Zhiyong Yuan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, China
| | - Yunchuan Sun
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, China.
| | - Lujun Zhao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, China.
| |
Collapse
|
20
|
Xie S, Zhang H, Liang Z, Yang X, Cao R. AXL, an Important Host Factor for DENV and ZIKV Replication. Front Cell Infect Microbiol 2021; 11:575346. [PMID: 33954117 PMCID: PMC8092360 DOI: 10.3389/fcimb.2021.575346] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Flaviviruses, as critically important pathogens, are still major public health problems all over the world. For instance, the evolution of ZIKV led to large-scale outbreaks in the Yap island in 2007. DENV was considered by the World Health Organization (WHO) as one of the 10 threats to global health in 2019. Enveloped viruses hijack a variety of host factors to complete its replication cycle. Phosphatidylserine (PS) receptor, AXL, is considered to be a candidate receptor for flavivirus invasion. In this review, we discuss the molecular structure of ZIKV and DENV, and how they interact with AXL to successfully invade host cells. A more comprehensive understanding of the molecular mechanisms of flavivirus-AXL interaction will provide crucial insights into the virus infection process and the development of anti-flavivirus therapeutics.
Collapse
Affiliation(s)
- Shengda Xie
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Huiru Zhang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhenjie Liang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xingmiao Yang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ruibing Cao
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
21
|
Rebelo R, Polónia B, Santos LL, Vasconcelos MH, Xavier CPR. Drug Repurposing Opportunities in Pancreatic Ductal Adenocarcinoma. Pharmaceuticals (Basel) 2021; 14:280. [PMID: 33804613 PMCID: PMC8003696 DOI: 10.3390/ph14030280] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is considered one of the deadliest tumors worldwide. The diagnosis is often possible only in the latter stages of the disease, with patients already presenting an advanced or metastatic tumor. It is also one of the cancers with poorest prognosis, presenting a five-year survival rate of around 5%. Treatment of PDAC is still a major challenge, with cytotoxic chemotherapy remaining the basis of systemic therapy. However, no major advances have been made recently, and therapeutic options are limited and highly toxic. Thus, novel therapeutic options are urgently needed. Drug repurposing is a strategy for the development of novel treatments using approved or investigational drugs outside the scope of the original clinical indication. Since repurposed drugs have already completed several stages of the drug development process, a broad range of data is already available. Thus, when compared with de novo drug development, drug repurposing is time-efficient, inexpensive and has less risk of failure in future clinical trials. Several repurposing candidates have been investigated in the past years for the treatment of PDAC, as single agents or in combination with conventional chemotherapy. This review gives an overview of the main drugs that have been investigated as repurposing candidates, for the potential treatment of PDAC, in preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Rita Rebelo
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (B.P.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Bárbara Polónia
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (B.P.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, IPO—Instituto Português de Oncologia, 4200-072 Porto, Portugal;
- ICBAS—Biomedical Sciences Institute Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - M. Helena Vasconcelos
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (B.P.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4200-135 Porto, Portugal
| | - Cristina P. R. Xavier
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (R.R.); (B.P.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4200-135 Porto, Portugal
| |
Collapse
|
22
|
Aehnlich P, Powell RM, Peeters MJW, Rahbech A, thor Straten P. TAM Receptor Inhibition-Implications for Cancer and the Immune System. Cancers (Basel) 2021; 13:cancers13061195. [PMID: 33801886 PMCID: PMC7998716 DOI: 10.3390/cancers13061195] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary TAM receptors are a family of receptor tyrosine kinases, comprising Tyro3, Axl and MerTK. Their primary role is in digestion of dying cells by macrophages without alarming the immune system. TAM receptors are also expressed by cancer cells in which signaling is oncogenic, and for this reason there is growing interest and research into TAM inhibition. This approach to cancer treatment may, however, come into conflict with beneficial and costimulatory TAM receptor signaling in T cells and natural killer (NK) cells. The aim of this review is to explore in detail the effects of TAM receptor inhibition on cancer cells and immune cells, and how the ramifications of this inhibition may affect cancer treatment in humans. Abstract Tyro3, Axl and MerTK (TAM) receptors are receptor tyrosine kinases which play important roles in efferocytosis and in the balancing of immune responses and inflammation. TAM receptor activation is induced upon binding of the ligands protein S (Pros1) or growth arrest-specific protein 6 (Gas6) which act as bridging molecules for binding of phosphatidyl serine (PtdSer) exposed on apoptotic cell membranes. Upon clearance of apoptotic cell material, TAM receptor activation on innate cells suppresses proinflammatory functions, thereby ensuring the immunologically silent removal of apoptotic material in the absence of deleterious immune responses. However, in T cells, MerTK signaling is costimulatory and promotes activation and functional output of the cell. MerTK and Axl are also aberrantly expressed in a range of both hematological and solid tumor malignancies, including breast, lung, melanoma and acute myeloid leukemia, where they have a role in oncogenic signaling. Consequently, TAM receptors are being investigated as therapeutic targets using small molecule inhibitors and have already demonstrated efficacy in mouse tumor models. Thus, inhibition of TAM signaling in cancer cells could have therapeutic value but given the opposing roles of TAM signaling in innate cells and T cells, TAM inhibition could also jeopardize anticancer immune responses. This conflict is discussed in this review, describing the effects of TAM inhibition on cancer cells as well as immune cells, while also examining the intricate interplay of cancer and immune cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Pia Aehnlich
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, 2730 Herlev, Denmark; (M.J.W.P.); (A.R.)
- Correspondence: (P.A.); (R.M.P.); (P.t.S.)
| | - Richard Morgan Powell
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, 2730 Herlev, Denmark; (M.J.W.P.); (A.R.)
- Correspondence: (P.A.); (R.M.P.); (P.t.S.)
| | - Marlies J. W. Peeters
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, 2730 Herlev, Denmark; (M.J.W.P.); (A.R.)
| | - Anne Rahbech
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, 2730 Herlev, Denmark; (M.J.W.P.); (A.R.)
| | - Per thor Straten
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, 2730 Herlev, Denmark; (M.J.W.P.); (A.R.)
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Correspondence: (P.A.); (R.M.P.); (P.t.S.)
| |
Collapse
|
23
|
Davra V, Kumar S, Geng K, Calianese D, Mehta D, Gadiyar V, Kasikara C, Lahey KC, Chang YJ, Wichroski M, Gao C, De Lorenzo MS, Kotenko SV, Bergsbaken T, Mishra PK, Gause WC, Quigley M, Spires TE, Birge RB. Axl and Mertk Receptors Cooperate to Promote Breast Cancer Progression by Combined Oncogenic Signaling and Evasion of Host Antitumor Immunity. Cancer Res 2021; 81:698-712. [PMID: 33239426 PMCID: PMC9999365 DOI: 10.1158/0008-5472.can-20-2066] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/23/2020] [Accepted: 11/19/2020] [Indexed: 11/16/2022]
Abstract
Despite the promising clinical benefit of targeted and immune checkpoint blocking therapeutics, current strategies have limited success in breast cancer, indicating that additional inhibitory pathways are required to complement existing therapeutics. TAM receptors (Tyro-3, Axl, and Mertk) are often correlated with poor prognosis because of their capacities to sustain an immunosuppressive environment. Here, we ablate Axl on tumor cells using CRISPR/Cas9 gene editing, and by targeting Mertk in the tumor microenvironment (TME), we observed distinct functions of TAM as oncogenic kinases, as well as inhibitory immune receptors. Depletion of Axl suppressed cell intrinsic oncogenic properties, decreased tumor growth, reduced the incidence of lung metastasis and increased overall survival of mice when injected into mammary fat pad of syngeneic mice, and demonstrated synergy when combined with anti-PD-1 therapy. Blockade of Mertk function on macrophages decreased efferocytosis, altered the cytokine milieu, and resulted in suppressed macrophage gene expression patterns. Mertk-knockout mice or treatment with anti-Mertk-neutralizing mAb also altered the cellular immune profile, resulting in a more inflamed tumor environment with enhanced T-cell infiltration into tumors and T-cell-mediated cytotoxicity. The antitumor activity from Mertk inhibition was abrogated by depletion of cytotoxic CD8α T cells by using anti-CD8α mAb or by transplantation of tumor cells into B6.CB17-Prkdc SCID mice. Our data indicate that targeting Axl expressed on tumor cells and Mertk in the TME is predicted to have a combinatorial benefit to enhance current immunotherapies and that Axl and Mertk have distinct functional activities that impair host antitumor response. SIGNIFICANCE: This study demonstrates how TAM receptors act both as oncogenic tyrosine kinases and as receptors that mediate immune evasion in cancer progression.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cells, Cultured
- Female
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immune Evasion/genetics
- Immune Evasion/immunology
- Immunotherapy/methods
- Kaplan-Meier Estimate
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/immunology
- Proto-Oncogene Proteins/metabolism
- Receptor Protein-Tyrosine Kinases/genetics
- Receptor Protein-Tyrosine Kinases/immunology
- Receptor Protein-Tyrosine Kinases/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- c-Mer Tyrosine Kinase/genetics
- c-Mer Tyrosine Kinase/immunology
- c-Mer Tyrosine Kinase/metabolism
- Axl Receptor Tyrosine Kinase
- Mice
Collapse
Affiliation(s)
- Viralkumar Davra
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Sushil Kumar
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Ke Geng
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - David Calianese
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Dhriti Mehta
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Varsha Gadiyar
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Canan Kasikara
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Kevin C Lahey
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Yun-Juan Chang
- Office of Advanced Research Computing, Rutgers- New Jersey Medical School, Newark, New Jersey
| | | | - Chan Gao
- Bristol Myers Squibb, Lawrenceville, New Jersey
| | | | - Sergei V Kotenko
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey
| | - Tessa Bergsbaken
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Pankaj K Mishra
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - William C Gause
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | | | | | - Raymond B Birge
- Department of Microbiology, Biochemistry and Molecular Genetics, Cancer Center, Rutgers- New Jersey Medical School, Newark, New Jersey.
| |
Collapse
|
24
|
Gadiyar V, Patel G, Davra V. Immunological role of TAM receptors in the cancer microenvironment. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 357:57-79. [PMID: 33234245 DOI: 10.1016/bs.ircmb.2020.09.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
TAM receptors belong to the family of receptor tyrosine kinases, comprising of Tyro3, Axl and Mertk receptors (TAMs) and are important homeostatic regulators of inflammation in higher eukaryotes. Along with their ligands, Gas6 and ProteinS, TAMs acts as receptors to phosphatidylserine (PtdSer), an anionic phospholipid that becomes externalized on the surface of apoptotic and stressed cells. TAM receptors, specially Mertk, have been well established to play a role in the process of efferocytosis, the engulfment of dying cells. Besides being efferocytic receptors, TAMs are pleiotropic immune modulators as the lack of TAM receptors in various mouse models lead to chronic inflammation and autoimmunity. Owing to their immune modulatory role, the PtdSer-TAM receptor signaling axis has been well characterized as a global immune-suppressive signal, and in cancers, and emerging literature implicates TAM receptors in cancer immunology and anti-tumor therapeutics. In the tumor microenvironment, immune-suppressive signals, such as ones that originate from TAM receptor signaling can be detrimental to anti-tumor therapy. In this chapter, we discuss immune modulatory functions of TAM receptors in the tumor microenvironment as well role of differentially expressed TAM receptors and their interactions with immune and tumor cells. Finally, we describe current strategies being utilized for targeting TAMs in several cancers and their implications in immunotherapy.
Collapse
Affiliation(s)
- Varsha Gadiyar
- Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Gopi Patel
- Rutgers New Jersey Medical School, Newark, NJ, United States
| | | |
Collapse
|
25
|
Chang W, Fa H, Xiao D, Wang J. Targeting phosphatidylserine for Cancer therapy: prospects and challenges. Theranostics 2020; 10:9214-9229. [PMID: 32802188 PMCID: PMC7415799 DOI: 10.7150/thno.45125] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is a leading cause of mortality and morbidity worldwide. Despite major improvements in current therapeutic methods, ideal therapeutic strategies for improved tumor elimination are still lacking. Recently, immunotherapy has attracted much attention, and many immune-active agents have been approved for clinical use alone or in combination with other cancer drugs. However, some patients have a poor response to these agents. New agents and strategies are needed to overcome such deficiencies. Phosphatidylserine (PS) is an essential component of bilayer cell membranes and is normally present in the inner leaflet. In the physiological state, PS exposure on the external leaflet not only acts as an engulfment signal for phagocytosis in apoptotic cells but also participates in blood coagulation, myoblast fusion and immune regulation in nonapoptotic cells. In the tumor microenvironment, PS exposure is significantly increased on the surface of tumor cells or tumor cell-derived microvesicles, which have innate immunosuppressive properties and facilitate tumor growth and metastasis. To date, agents targeting PS have been developed, some of which are under investigation in clinical trials as combination drugs for various cancers. However, controversial results are emerging in laboratory research as well as in clinical trials, and the efficiency of PS-targeting agents remains uncertain. In this review, we summarize recent progress in our understanding of the physiological and pathological roles of PS, with a focus on immune suppressive features. In addition, we discuss current drug developments that are based on PS-targeting strategies in both experimental and clinical studies. We hope to provide a future research direction for the development of new agents for cancer therapy.
Collapse
Affiliation(s)
- Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
| | - Hongge Fa
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| | - Dandan Xiao
- Institute for Translational Medicine, The Affiliated Hospital, College of medicine, Qingdao University, Qingdao, China
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| | - Jianxun Wang
- School of Basic Medical Sciences, College of medicine, Qingdao University, Qingdao, China
| |
Collapse
|