101
|
Regulatory (FoxP3 +) T cells and TGF-β predict the response to anti-PD-1 immunotherapy in patients with non-small cell lung cancer. Sci Rep 2020; 10:18994. [PMID: 33149213 PMCID: PMC7642363 DOI: 10.1038/s41598-020-76130-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/01/2020] [Indexed: 01/28/2023] Open
Abstract
Antitumor immune responses induced by immune checkpoint inhibitors anti-PD-1 or anti-PD-L1 have been used as therapeutic strategies in advanced non-small cell lung cancer (NSCLC) patients over the last decade. Favorable antitumor activity to immune checkpoint inhibitors is correlated with high PD-L1 expression, increased tumor-infiltrating lymphocytes, and decreased suppressive immune cells including Treg cells, myeloid-derived suppressor cells, or tumor-associated macrophages in various cancer types. In this study, we investigated the potential correlation between clinical outcomes and peripheral blood immune cell profiles, specifically focused on FoxP3+ Treg cells, collected at baseline and one week after anti-PD-1 therapy in two independent cohorts of patients with NSCLC: a discovery cohort of 83 patients and a validation cohort of 49 patients. High frequencies of circulating Treg cells one week after anti-PD-1 therapy were correlated with a high response rate, longer progression-free survival, and overall survival. Furthermore, high levels of TGF-β and Treg cells were associated with favorable clinical outcomes. Our results suggest that higher levels of FoxP3+ Treg cells and TGF-β can predict a favorable response to anti-PD-1 immunotherapy in patients with advanced NSCLC.
Collapse
|
102
|
Wang X, Wu Z, Qiu W, Chen P, Xu X, Han W. Programming CAR T cells to enhance anti-tumor efficacy through remodeling of the immune system. Front Med 2020; 14:726-745. [PMID: 32794014 DOI: 10.1007/s11684-020-0746-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor (CAR) T cells have been indicated effective in treating B cell acute lymphoblastic leukemia and non-Hodgkin lymphoma and have shown encouraging results in preclinical and clinical studies. However, CAR T cells have achieved minimal success against solid malignancies because of the additional obstacles of their insufficient migration into tumors and poor amplification and persistence, in addition to antigen-negative relapse and an immunosuppressive microenvironment. Various preclinical studies are exploring strategies to overcome the above challenges. Mobilization of endogenous immune cells is also necessary for CAR T cells to obtain their optimal therapeutic effect given the importance of the innate immune responses in the elimination of malignant tumors. In this review, we focus on the recent advances in the engineering of CAR T cell therapies to restore the immune response in solid malignancies, especially with CAR T cells acting as cellular carriers to deliver immunomodulators to tumors to mobilize the endogenous immune response. We also explored the sensitizing effects of conventional treatment approaches, such as chemotherapy and radiotherapy, on CAR T cell therapy. Finally, we discuss the combination of CAR T cells with biomaterials or oncolytic viruses to enhance the anti-tumor outcomes of CAR T cell therapies in solid tumors.
Collapse
Affiliation(s)
- Xiaohui Wang
- College of Biotechnology, Southwest University, Chongqing, 400715, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China.,Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhiqiang Wu
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei Qiu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China
| | - Ping Chen
- College of Biotechnology, Southwest University, Chongqing, 400715, China
| | - Xiang Xu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China.
| | - Weidong Han
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
103
|
Giri PS, Dwivedi M, Begum R. Decreased suppression of CD8 + and CD4 + T cells by peripheral regulatory T cells in generalized vitiligo due to reduced NFATC1 and FOXP3 proteins. Exp Dermatol 2020; 29:759-775. [PMID: 32682346 DOI: 10.1111/exd.14157] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/30/2020] [Accepted: 07/11/2020] [Indexed: 02/05/2023]
Abstract
Regulatory T cells (Tregs) are involved in the suppression of activated T cells in generalized vitiligo (GV). The study was aimed to investigate Tregs functional defects in Treg:CD8+ and Treg:CD4+ T cells' co-culture systems of 55 GV patients and 45 controls. CD8+ and CD4+ T-cell proliferation was assessed by BrdU assay; production of IL-10, TGF-β and IFN-γ cytokines was assessed by ELISA; and FOXP3, CD25, NFATC1 and CD44 proteins were measured by flow cytometry. Generalized vitiligo patients showed reduced suppression of CD8+ and CD4+ T cells (P = .0384, P = .0084), increased IFN-γ (P < .0001, P = .0019), decreased IL-10 and TGF-β (P < .0001) and decreased FOXP3, CD25 and NFATC1 proteins (P < .0001). Active vitiligo (AV) patients showed reduced suppression of CD8+ & CD4+ T cells (P = .006, P = .015), increased IFN-γ (P = .036, P = .045), decreased IL-10 (P = .009, P = .021), FOXP3 (P = .0244) and NFATC1 (P = .019). Severe GV (50%-75% VASI) patients showed reduced suppression of CD8+ and CD4+ T cells (P = .0003, P = .001), increased IFN-γ (P = .0029, P < .0001), decreased IL-10 (P = .0057, P = .0017), FOXP3 (P = .002) and NFATC1 (P = .0347). VASI score was positively correlated with the suppression of CD8+ and CD4+ T cells (P = .0006, P < .0001), IL-10 (P = .0096, P = .029), FOXP3 (P = .0008) and NFATC1 (P = .043), whereas it was negatively correlated with IFN-γ (P = .0029, P = .0017). Early age of onset patients' Tregs demonstrated decreased suppression of CD8+ and CD4+ T cells (P = .0156, P = .0074), decreased TGF-β (P = .0212, P = .0083) and NFATC1 (P = .0103). NFATC1 was positively correlated with FOXP3 in Tregs (P < .0001). Our results suggest impaired Tregs suppressive function in GV patients due to decreased NFATC1, FOXP3, CD25, IL-10 and TGF-β resulting into increased CD8+ and CD4+ T-cell proliferation and IFN-γ production. For the first time, decreased NFATC1 levels were correlated with decreased FOXP3, thereby altering Treg cell function in GV patients. Additionally, decreased Treg cell function also affected onset, activity and severity of GV.
Collapse
Affiliation(s)
- Prashant S Giri
- Faculty of Science, C. G. Bhakta Institute of Biotechnology, Uka Tarsadia University, Surat, India
| | - Mitesh Dwivedi
- Faculty of Science, C. G. Bhakta Institute of Biotechnology, Uka Tarsadia University, Surat, India
| | - Rasheedunnisa Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
104
|
Jo Y, Ali LA, Shim JA, Lee BH, Hong C. Innovative CAR-T Cell Therapy for Solid Tumor; Current Duel between CAR-T Spear and Tumor Shield. Cancers (Basel) 2020; 12:cancers12082087. [PMID: 32731404 PMCID: PMC7464778 DOI: 10.3390/cancers12082087] [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: 06/09/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Novel engineered T cells containing chimeric antigen receptors (CAR-T cells) that combine the benefits of antigen recognition and T cell response have been developed, and their effect in the anti-tumor immunotherapy of patients with relapsed/refractory leukemia has been dramatic. Thus, CAR-T cell immunotherapy is rapidly emerging as a new therapy. However, it has limitations that prevent consistency in therapeutic effects in solid tumors, which accounts for over 90% of all cancer patients. Here, we review the literature regarding various obstacles to CAR-T cell immunotherapy for solid tumors, including those that cause CAR-T cell dysfunction in the immunosuppressive tumor microenvironment, such as reactive oxygen species, pH, O2, immunosuppressive cells, cytokines, and metabolites, as well as those that impair cell trafficking into the tumor microenvironment. Next-generation CAR-T cell therapy is currently undergoing clinical trials to overcome these challenges. Therefore, novel approaches to address the challenges faced by CAR-T cell immunotherapy in solid tumors are also discussed here.
Collapse
Affiliation(s)
- Yuna Jo
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Laraib Amir Ali
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Ju A Shim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Byung Ha Lee
- NeoImmuneTech, Inc., 2400 Research Blvd., Suite 250, Rockville, MD 20850, USA;
| | - Changwan Hong
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
- Correspondence: ; Tel.: +82-51-510-8041
| |
Collapse
|
105
|
Derynck R, Turley SJ, Akhurst RJ. TGFβ biology in cancer progression and immunotherapy. Nat Rev Clin Oncol 2020; 18:9-34. [DOI: 10.1038/s41571-020-0403-1] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2020] [Indexed: 02/07/2023]
|
106
|
Takeda Y, Kobayashi S, Kitakaze M, Yamada D, Akita H, Asai A, Konno M, Arai T, Kitagawa T, Ofusa K, Yabumoto M, Hirotsu T, Vecchione A, Taniguchi M, Doki Y, Eguchi H, Ishii H. Immuno-Surgical Management of Pancreatic Cancer with Analysis of Cancer Exosomes. Cells 2020; 9:cells9071645. [PMID: 32659892 PMCID: PMC7408222 DOI: 10.3390/cells9071645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/05/2020] [Accepted: 07/05/2020] [Indexed: 02/07/2023] Open
Abstract
Exosomes (EXs), a type of extracellular vesicles secreted from various cells and especially cancer cells, mesenchymal cells, macrophages and other cells in the tumor microenvironment (TME), are involved in biologically malignant behaviors of cancers. Recent studies have revealed that EXs contain microRNAs on their inside and express proteins and glycolipids on their outsides, every component of which plays a role in the transmission of genetic and/or epigenetic information in cell-to-cell communications. It is also known that miRNAs are involved in the signal transduction. Thus, EXs may be useful for monitoring the TME of tumor tissues and the invasion and metastasis, processes that are associated with patient survival. Because several solid tumors secrete immune checkpoint proteins, including programmed cell death-ligand 1, the EX-mediated mechanisms are suggested to be potent targets for monitoring patients. Therefore, a companion therapeutic approach against cancer metastasis to distant organs is proposed when surgical removal of the primary tumor is performed. However, EXs and immune checkpoint mechanisms in pancreatic cancer are not fully understood, we provide an update on the recent advances in this field and evidence that EXs will be useful for maximizing patient benefit in precision medicine.
Collapse
Affiliation(s)
- Yu Takeda
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Masatoshi Kitakaze
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Hirofumi Akita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Ayumu Asai
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Artificial Intelligence Research Center, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan;
| | - Masamitsu Konno
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Takahiro Arai
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Unitech Co., Ltd., Kashiwa 277-0005, Japan
| | - Toru Kitagawa
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
- Kyowa-kai Medical Corporation, Osaka 540-0008, Japan
| | - Ken Ofusa
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Prophoenix Division, Food and Life-Science Laboratory, Idea Consultants, Inc., Osaka-city, Osaka 559-8519, Japan
| | - Masami Yabumoto
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
- Kinshu-kai Medical Corporation, Osaka 558-0041, Japan
| | - Takaaki Hirotsu
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Hirotsu Bio Science Inc., Tokyo 107-0062, Japan
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, University of Rome “Sapienza”, Santo Andrea Hospital, via di Grottarossa, 1035-00189 Rome, Italy;
| | - Masateru Taniguchi
- Artificial Intelligence Research Center, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan;
| | - Yuichiro Doki
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Hidetoshi Eguchi
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Hideshi Ishii
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
- Correspondence: ; Tel.: +81-(0)6-6210-8406 (ext. 8405); Fax: +81-(0)6-6210-8407
| |
Collapse
|
107
|
Ciner AT, Jones K, Muschel RJ, Brodt P. The unique immune microenvironment of liver metastases: Challenges and opportunities. Semin Cancer Biol 2020; 71:143-156. [PMID: 32526354 DOI: 10.1016/j.semcancer.2020.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 02/07/2023]
Abstract
Liver metastases from gastrointestinal and non-gastrointestinal malignancies remain a major cause of cancer-related mortality and a major clinical challenge. The liver has unique properties that facilitate metastatic expansion, including a complex immune system that evolved to dampen immunity to neoantigens entering the liver from the gut, through the portal circulation. In this review, we describe the unique microenvironment encountered by cancer cells in the liver, focusing on elements of the innate and adaptive immune response that can act as a double-edge sword, contributing to the elimination of cancer cells on the one hand and promoting their survival and growth, on the other. We discuss this microenvironment in a clinical context, particularly for colorectal carcinoma, and highlight how a better understanding of the role of the microenvironment has spurred an intense effort to develop novel and innovative strategies for targeting liver metastatic disease, some of which are currently being tested in the clinic.
Collapse
Affiliation(s)
- Aaron T Ciner
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Keaton Jones
- Oxford Institute for Radiation Oncology, Department of Surgery, University of Oxford, Oxford, UK
| | - Ruth J Muschel
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Pnina Brodt
- Departments of Surgery, Medicine and Oncology, McGill University, and the Research Institute of the McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
108
|
Renin-Angiotensin System in Lung Tumor and Microenvironment Interactions. Cancers (Basel) 2020; 12:cancers12061457. [PMID: 32503281 PMCID: PMC7352181 DOI: 10.3390/cancers12061457] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/24/2020] [Accepted: 06/01/2020] [Indexed: 02/08/2023] Open
Abstract
The mechanistic involvement of the renin-angiotensin system (RAS) reaches beyond cardiovascular physiopathology. Recent knowledge pinpoints a pleiotropic role for this system, particularly in the lung, and mainly through locally regulated alternative molecules and secondary pathways. Angiotensin peptides play a role in cell proliferation, immunoinflammatory response, hypoxia and angiogenesis, which are critical biological processes in lung cancer. This manuscript reviews the literature supporting a role for the renin-angiotensin system in the lung tumor microenvironment and discusses whether blockade of this pathway in clinical settings may serve as an adjuvant therapy in lung cancer.
Collapse
|
109
|
Wang J, Matosevic S. Functional and metabolic targeting of natural killer cells to solid tumors. Cell Oncol (Dordr) 2020; 43:577-600. [DOI: 10.1007/s13402-020-00523-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2020] [Indexed: 12/15/2022] Open
|
110
|
Menon N, Dang HX, Datla US, Moarefian M, Lawrence CB, Maher CA, Jones CN. Heparin-based hydrogel scaffolding alters the transcriptomic profile and increases the chemoresistance of MDA-MB-231 triple-negative breast cancer cells. Biomater Sci 2020; 8:2786-2796. [PMID: 32091043 PMCID: PMC7497406 DOI: 10.1039/c9bm01481k] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 02/08/2020] [Indexed: 12/30/2022]
Abstract
The tumor microenvironment plays a critical role in the proliferation and chemoresistance of cancer cells. Growth factors (GFs) are known to interact with the extracellular matrix (ECM) via heparin binding sites, and these associations influence cell behavior. In the present study, we demonstrate the ability to define signals presented by the scaffold by pre-mixing growth factors, such as epidermal growth factor, into the heparin-based (HP-B) hydrogel prior to gelation. In the 3D biomimetic microenvironment, breast cancer cells formed spheroids within 24 hours of initial seeding. Despite higher number of proliferating cells in 2D cultures, 3D spheroids exhibited a higher degree of chemoresistance after 72 hours. Further, our RNA sequencing results highlighted the phenotypic changes influenced by solid-phase GF presentation. Wnt/β-catenin and TGF-β signaling were upregulated in the cells grown in the hydrogel, while apoptosis, IL2-STAT5 and PI3K-AKT-mTOR signaling were downregulated. With emerging technologies for precision medicine in cancer, this nature of fine-tuning the microenvironment is paramount for cultivation and downstream characterization of primary cancer cells and rare circulating tumor cells (CTCs), and effective screening of chemotherapeutic agents.
Collapse
Affiliation(s)
- Nidhi Menon
- Graduate Program in Translational Biology
, Medicine and Health
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
.
- Department of Biological Sciences
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
| | - Ha X. Dang
- McDonnell Genome Institute
, Washington University in St. Louis
,
MO
63108
, USA
- Department of Medicine
, Washington University School of Medicine
,
St. Louis
, MO
63108
, USA
- Alvin J. Siteman Cancer Center
, Washington University in St. Louis
,
St. Louis
, MO
63108
, USA
| | - Udaya Sree Datla
- Graduate Program in Translational Biology
, Medicine and Health
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
.
- Department of Biological Sciences
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
| | - Maryam Moarefian
- Department of Mechanical Engineering
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
| | - Christopher B. Lawrence
- Department of Biological Sciences
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
| | - Christopher A. Maher
- McDonnell Genome Institute
, Washington University in St. Louis
,
MO
63108
, USA
- Department of Medicine
, Washington University School of Medicine
,
St. Louis
, MO
63108
, USA
- Alvin J. Siteman Cancer Center
, Washington University in St. Louis
,
St. Louis
, MO
63108
, USA
- Department of Biomedical Engineering
, Washington University in St. Louis
,
MO
63108
, USA
| | - Caroline N. Jones
- Graduate Program in Translational Biology
, Medicine and Health
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
.
- Department of Biological Sciences
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
| |
Collapse
|
111
|
Roth TL, Li PJ, Blaeschke F, Nies JF, Apathy R, Mowery C, Yu R, Nguyen MLT, Lee Y, Truong A, Hiatt J, Wu D, Nguyen DN, Goodman D, Bluestone JA, Ye CJ, Roybal K, Shifrut E, Marson A. Pooled Knockin Targeting for Genome Engineering of Cellular Immunotherapies. Cell 2020; 181:728-744.e21. [PMID: 32302591 PMCID: PMC7219528 DOI: 10.1016/j.cell.2020.03.039] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/13/2020] [Accepted: 03/18/2020] [Indexed: 12/12/2022]
Abstract
Adoptive transfer of genetically modified immune cells holds great promise for cancer immunotherapy. CRISPR knockin targeting can improve cell therapies, but more high-throughput methods are needed to test which knockin gene constructs most potently enhance primary cell functions in vivo. We developed a widely adaptable technology to barcode and track targeted integrations of large non-viral DNA templates and applied it to perform pooled knockin screens in primary human T cells. Pooled knockin of dozens of unique barcoded templates into the T cell receptor (TCR)-locus revealed gene constructs that enhanced fitness in vitro and in vivo. We further developed pooled knockin sequencing (PoKI-seq), combining single-cell transcriptome analysis and pooled knockin screening to measure cell abundance and cell state ex vivo and in vivo. This platform nominated a novel transforming growth factor β (TGF-β) R2-41BB chimeric receptor that improved solid tumor clearance. Pooled knockin screening enables parallelized re-writing of endogenous genetic sequences to accelerate discovery of knockin programs for cell therapies.
Collapse
Affiliation(s)
- Theodore L Roth
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA.
| | - P Jonathan Li
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Franziska Blaeschke
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Jasper F Nies
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Ryan Apathy
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Cody Mowery
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Ruby Yu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Michelle L T Nguyen
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Youjin Lee
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Anna Truong
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Joseph Hiatt
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - David Wu
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - David N Nguyen
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel Goodman
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Jeffrey A Bluestone
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA, USA
| | - Chun Jimmie Ye
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA; Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Institute of Computational Health Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Kole Roybal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA; Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA, USA; UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Eric Shifrut
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Alexander Marson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA; UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
112
|
Skeik N, Golden M, Berg A, North M. Type A Aortic Dissection Caused by Loeys-Dietz Syndrome with Novel Variation. Ann Vasc Surg 2020; 68:567.e1-567.e4. [PMID: 32339686 DOI: 10.1016/j.avsg.2020.04.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/07/2020] [Indexed: 11/16/2022]
Abstract
Loeys-Dietz syndrome (LDS) is a rare autosomal-dominant connective tissue disorder that can lead to aortic aneurysm and dissection. There are 5 different types caused by mutations in TGFβR1 (transforming growth factor β receptor), TGFβR2, SMAD3, TGFβ2 (transforming growth factor β), and TGFβ3 respectively. The prevalence of LDS is estimated to be less than 1 in 100,000. There is considerable variability in the phenotype of LDS, from mild features to severe systemic abnormalities. There is overlap in the manifestations of LDS and Marfan syndrome, including increased risk of ascending aortic aneurysm and aortic dissection, as well as abnormally long limbs and fingers. Management can be very challenging with a high risk of complications with revascularization. We report a 60-year-old female who presented with a type A aortic dissection that originated from the aortic root and extended to the bilateral common femoral arteries. Genetic testing revealed a novel alteration of the TGFβR1 gene (c689 C>A in exon 4) that to our knowledge has not been previously reported or found in large population cohorts. She was managed through a Bentall procedure that was complicated by a graft tear and stenosis of the distal anastomosis site, in addition to requiring a temporary pacemaker implantation and hemodialysis after the procedure. Ultimately, the patient was able to recover fully.
Collapse
Affiliation(s)
- Nedaa Skeik
- Department of Vascular Medicine, Minneapolis Heart Institute at Abbott Northwestern Hospital, Minneapolis, MN.
| | - Max Golden
- Research Department, Minneapolis Heart Institute Foundation, Minneapolis, MN
| | - Allison Berg
- Genetic Department, Children's Minnesota Genetics, Minneapolis, MN
| | - Monique North
- Research Department, Minneapolis Heart Institute Foundation, Minneapolis, MN
| |
Collapse
|
113
|
Manuszak C, Brainard M, Thrash E, Hodi FS, Severgnini M. Standardized 11-color flow cytometry panel for the functional phenotyping of human T regulatory cells. J Biol Methods 2020; 7:e131. [PMID: 32313815 PMCID: PMC7163208 DOI: 10.14440/jbm.2020.325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/02/2020] [Accepted: 03/21/2020] [Indexed: 12/17/2022] Open
Abstract
T regulatory cells (Tregs) are a cell subset that can suppress immune responses to maintain homeostasis and self-tolerance. In some scenarios, the immunosuppressive nature could be associated to other pathological developments such as autoimmune diseases and cancers. Due to the importance of Tregs in disease pathogenesis, we developed and validated an 11-color flow cytometry panel for phenotypic and functional detection of Treg markers using healthy human donor peripheral blood mononuclear cells (PBMCs). Our panel contains 4 Treg surface proteins and 2 functional cytokines as well as T-lymphocyte lineage markers CD3, CD4, and CD8. Our data shows an increase in expression of markers CD25, FoxP3, CTLA4, GITR and intracellular cytokines IL4 and TGFβ when comparing unstimulated samples to CD3/CD28 bead stimulated samples. This 11-color panel can be used to functionally evaluate immunosuppressive Tregs in human PBMC samples.
Collapse
Affiliation(s)
- Claire Manuszak
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline, Ave Mayer Building 305, Boston, MA 02215, USA
| | - Martha Brainard
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline, Ave Mayer Building 305, Boston, MA 02215, USA
| | - Emily Thrash
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline, Ave Mayer Building 305, Boston, MA 02215, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline, Ave Mayer Building 305, Boston, MA 02215, USA
| | - Mariano Severgnini
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline, Ave Mayer Building 305, Boston, MA 02215, USA
| |
Collapse
|
114
|
Larson C, Oronsky B, Carter CA, Oronsky A, Knox SJ, Sher D, Reid TR. TGF-beta: a master immune regulator. Expert Opin Ther Targets 2020; 24:427-438. [PMID: 32228232 DOI: 10.1080/14728222.2020.1744568] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Transforming Growth Factor-Beta (TGF-β) is a master regulator of numerous cellular functions including cellular immunity. In cancer, TGF-β can function as a tumor promoter via several mechanisms including immunosuppression. Since the immune checkpoint pathways are co-opted in cancer to induce T cell tolerance, this review posits that TGF-β is a master checkpoint in cancer, whose negative regulatory influence overrides and controls that of other immune checkpoints.Areas Covered: This review examines therapeutic agents that target TGF-β and its signaling pathways for the treatment of cancer which may be classifiable as checkpoint inhibitors in the broadest sense. This concept is supported by the observations that 1) only a subset of patients benefit from current checkpoint inhibitor therapies, 2) the presence of TGF-β in the tumor microenvironment is associated with excluded or cold tumors, and resistance to checkpoint inhibitors, and 3) existing biomarkers such as PD-1, PD-L1, microsatellite instability and tumor mutational burden are inadequate to reliably and adequately identify immuno-responsive patients. By contrast, TGF-β overexpression is a widespread and profoundly negative molecular hallmark in multiple tumor types.Expert Opinion: TGF-β status may serve as a biomarker to predict responsiveness and as a therapeutic target to increase the activity of immunotherapies.
Collapse
Affiliation(s)
| | | | | | - Arnold Oronsky
- EpicentRx, San Diego, CA, USA.,InterWest Partners, Menlo Park, CA, USA
| | - Susan J Knox
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - David Sher
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Tony R Reid
- Department of Medical Oncology, UC San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
115
|
Uhl LFK, Gérard A. Modes of Communication between T Cells and Relevance for Immune Responses. Int J Mol Sci 2020; 21:E2674. [PMID: 32290500 PMCID: PMC7215318 DOI: 10.3390/ijms21082674] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 11/16/2022] Open
Abstract
T cells are essential mediators of the adaptive immune system, which constantly patrol the body in search for invading pathogens. During an infection, T cells that recognise the pathogen are recruited, expand and differentiate into subtypes tailored to the infection. In addition, they differentiate into subsets required for short and long-term control of the pathogen, i.e., effector or memory. T cells have a remarkable degree of plasticity and heterogeneity in their response, however, their overall response to a given infection is consistent and robust. Much research has focused on how individual T cells are activated and programmed. However, in order to achieve a critical level of population-wide reproducibility and robustness, neighbouring cells and surrounding tissues have to provide or amplify relevant signals to tune the overall response accordingly. The characteristics of the immune response-stochastic on the individual cell level, robust on the global level-necessitate coordinated responses on a system-wide level, which facilitates the control of pathogens, while maintaining self-tolerance. This global coordination can only be achieved by constant cellular communication between responding cells, and faults in this intercellular crosstalk can potentially lead to immunopathology or autoimmunity. In this review, we will discuss how T cells mount a global, collective response, by describing the modes of T cell-T cell (T-T) communication they use and highlighting their physiological relevance in programming and controlling the T cell response.
Collapse
Affiliation(s)
| | - Audrey Gérard
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK;
| |
Collapse
|
116
|
Messex JK, Byrd CJ, Liou GY. Signaling of Macrophages that Contours the Tumor Microenvironment for Promoting Cancer Development. Cells 2020; 9:cells9040919. [PMID: 32283687 PMCID: PMC7226754 DOI: 10.3390/cells9040919] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
The immune response is critical in the maintenance of an organism’s health. The immune response can be broken down into two groups. The innate response, which is fast-acting and rids the body of most foreign material before infection occurs, and the adaptive response, a more specific defense against pathogen composed mostly of antibody production and killer cells. Linking the two responses via cytokine and chemokine secretion are macrophages, motile phagocytic cells that ingest and present foreign material playing a role in the innate and adaptive immune response. Although macrophages are necessary for the survival of an organism, studies have also shown macrophages play a more sinister role in the initiation, progression, and metastasis in tumorous cells. In this comprehensive review, we show how macrophages induce such a response through abnormal cellular signaling and creating a cellular microenvironment conducive for tumor growth and metastasis, as well as the future outlook of this field.
Collapse
Affiliation(s)
- Justin K. Messex
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA; (J.K.M.); (C.J.B.)
| | - Crystal J. Byrd
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA; (J.K.M.); (C.J.B.)
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Geou-Yarh Liou
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA; (J.K.M.); (C.J.B.)
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
- Correspondence: ; Tel.: +1-(404)-880-6981; Fax: +1-(404)-880-6756
| |
Collapse
|
117
|
Mondini M, Levy A, Meziani L, Milliat F, Deutsch E. Radiotherapy-immunotherapy combinations - perspectives and challenges. Mol Oncol 2020; 14:1529-1537. [PMID: 32112478 PMCID: PMC7332212 DOI: 10.1002/1878-0261.12658] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/18/2019] [Accepted: 02/27/2020] [Indexed: 12/23/2022] Open
Abstract
Ionizing radiation has historically been used to treat cancer by killing tumour cells, in particular by inducing DNA damage. This view of radiotherapy (RT) as a simple cytotoxic agent has dramatically changed in recent years, and it is now widely accepted that RT can deeply reshape the tumour environment by modulating the immune response. Such evidence gives a strong rationale for the use of immunomodulators to boost the therapeutic value of RT, introducing the era of ‘immunoradiotherapy’. The increasing amount of preclinical and clinical data concerning the combination of RT with immunomodulators, in particular with immune checkpoint inhibitors such as anti‐PD‐1/PD‐L1 and anti‐CTLA4, reflects the interest of the scientific and medical community concerning immunoradiotherapy. The expectations are enormous since the rationale for performing such combinations is strong, with the possibility to use a local treatment such as RT to amplify a systemic antitumour response, as illustrated by the case of the abscopal effect. Nevertheless, several points remain to be addressed such as the need to find biomarkers to identify patients who will benefit from immunoradiotherapy, the identification of the best sequences/schedules for combination with immunomodulators and mechanisms to overcome resistance. Additionally, the effects of immunoradiotherapy on healthy tissues and related toxicity remain largely unexplored. To answer these critical questions and make immunoradiotherapy keep its promising qualities, large efforts are needed from both the pharmaceutical industry and academic/governmental research. Moreover, because of the work of both these entities, the arsenal of available immunomodulators is quickly expanding, thus opening the field to increasing combinations with RT. We thus forecast that the field of immunoradiotherapy will further expand in the coming years, and it needs to be supported by appropriate investment plans.
Collapse
Affiliation(s)
- Michele Mondini
- Gustave Roussy, Université Paris-Saclay, SIRIC SOCRATE, Villejuif, France.,INSERM, U1030, Labex LERMIT, Villejuif, France
| | - Antonin Levy
- INSERM, U1030, Labex LERMIT, Villejuif, France.,Département de Radiothérapie, Gustave Roussy, Université Paris-Saclay, DHU TORINO, Villejuif, France
| | - Lydia Meziani
- Gustave Roussy, Université Paris-Saclay, SIRIC SOCRATE, Villejuif, France.,INSERM, U1030, Labex LERMIT, Villejuif, France
| | - Fabien Milliat
- Department of Radiobiology and regenerative Medicine (SERAMED), Laboratory of Medical Radiobiology (LRMed), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Eric Deutsch
- Gustave Roussy, Université Paris-Saclay, SIRIC SOCRATE, Villejuif, France.,INSERM, U1030, Labex LERMIT, Villejuif, France.,Département de Radiothérapie, Gustave Roussy, Université Paris-Saclay, DHU TORINO, Villejuif, France
| |
Collapse
|
118
|
Tang N, Cheng C, Zhang X, Qiao M, Li N, Mu W, Wei XF, Han W, Wang H. TGF-β inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors. JCI Insight 2020. [PMID: 31999649 DOI: 10.1172/jci.insight.133977.pmid:31999649;pmcid:pmc7101140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
In recent years, chimeric antigen receptor-modified T cell (CAR T cell) therapy has proven to be a promising approach against cancer. Nonetheless, this approach still faces multiple challenges in eliminating solid tumors, one of which being the immunosuppressive tumor microenvironment (TME). Here, we demonstrated that knocking out the endogenous TGF-β receptor II (TGFBR2) in CAR T cells with CRISPR/Cas9 technology could reduce the induced Treg conversion and prevent the exhaustion of CAR T ce lls. Meanwhile, TGFBR2-edited CAR T cells had better in vivo tumor elimination efficacy, both in cell line-derived xenograft and patient-derived xenograft solid tumor models, whether administered locally or systemically. In addition, the TGFBR2-edited CAR T cells could eliminate contralaterally reinoculated xenografts in mice effectively, with an increased proportion of memory subsets within circulating CAR T cells of central memory and effector memory subsets. In conclusion, we greatly improved the in vitro and in vivo function of CAR T cells in TGF-β-rich tumor environments by knocking out endogenous TGFBR2 and propose a potentially new method to improve the efficacy of CAR T cell therapy for treating solid tumors.
Collapse
Affiliation(s)
- Na Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chen Cheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Xingying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Miaomiao Qiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Na Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei Mu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | - Weidong Han
- Biotherapeutic Department and
- Department of Molecular Biology and Immunology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Haoyi Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
119
|
Tang N, Cheng C, Zhang X, Qiao M, Li N, Mu W, Wei XF, Han W, Wang H. TGF-β inhibition via CRISPR promotes the long-term efficacy of CAR T cells against solid tumors. JCI Insight 2020; 5:133977. [PMID: 31999649 DOI: 10.1172/jci.insight.133977] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/15/2020] [Indexed: 12/21/2022] Open
Abstract
In recent years, chimeric antigen receptor-modified T cell (CAR T cell) therapy has proven to be a promising approach against cancer. Nonetheless, this approach still faces multiple challenges in eliminating solid tumors, one of which being the immunosuppressive tumor microenvironment (TME). Here, we demonstrated that knocking out the endogenous TGF-β receptor II (TGFBR2) in CAR T cells with CRISPR/Cas9 technology could reduce the induced Treg conversion and prevent the exhaustion of CAR T ce lls. Meanwhile, TGFBR2-edited CAR T cells had better in vivo tumor elimination efficacy, both in cell line-derived xenograft and patient-derived xenograft solid tumor models, whether administered locally or systemically. In addition, the TGFBR2-edited CAR T cells could eliminate contralaterally reinoculated xenografts in mice effectively, with an increased proportion of memory subsets within circulating CAR T cells of central memory and effector memory subsets. In conclusion, we greatly improved the in vitro and in vivo function of CAR T cells in TGF-β-rich tumor environments by knocking out endogenous TGFBR2 and propose a potentially new method to improve the efficacy of CAR T cell therapy for treating solid tumors.
Collapse
Affiliation(s)
- Na Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chen Cheng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Xingying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Miaomiao Qiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Na Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei Mu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | | | - Weidong Han
- Biotherapeutic Department and.,Department of Molecular Biology and Immunology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Haoyi Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
120
|
Foughali R, Marie JC. [TGF-β-targeted immunotherapy aiming for new and efficient treatments of cancer]. Med Sci (Paris) 2020; 36:77-79. [PMID: 32014103 DOI: 10.1051/medsci/2019261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Rayan Foughali
- Master de cancérologie, ISPB, Université Claude Bernard Lyon 1, Lyon 69008, France
| | - Julien C Marie
- Tumor escape resistance immunity department, Cancer research center of Lyon, Lyon 69008, France
| |
Collapse
|
121
|
Zhang G, Lu J, Yang M, Wang Y, Liu H, Xu C. Elevated GALNT10 expression identifies immunosuppressive microenvironment and dismal prognosis of patients with high grade serous ovarian cancer. Cancer Immunol Immunother 2020; 69:175-187. [PMID: 31853576 DOI: 10.1007/s00262-019-02454-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 12/09/2019] [Indexed: 01/07/2023]
Abstract
High grade ovarian serous cancer (HGSC) is a malignant disease with high mortality. Glycosylation plays important roles in tumor invasion and immune evasion, but its effect on the immune microenvironment of HGSC remains unclear. This study examined the association of glycosyltransferase expression with HGSC prognosis and explored the underlying mechanism using clinical specimens and integrated bioinformatic analyses. We identified a cluster of 15 glycogenes associated with reduced overall survival, and GALNT10 was found to be an independent predictor of HGSC prognosis. The high GALNT10 expression was associated with increased regulatory CD4+ T cells infiltration and decreased granzyme B expression in CD8+ T cells. The expression of GALNT10 and its product, Tn antigen, in HGSC specimens was associated with the increased infiltration of M2 macrophages and neutrophils, and the decreased infiltration of CD3+ T cells, NK cells, and B cells. Taken collectively, high GALNT10 expression confers with immunosuppressive microenvironment to promote tumor progression and predicts poor clinical outcomes in HGSC patients.
Collapse
Affiliation(s)
- Guodong Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, No. 128, Shenyang Road, Shanghai, 200011, China
| | - Jiaqi Lu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Department of Gynecology, Kashgar Prefecture Second People's Hospital, Kashi, Xinjiang, 844000, China
| | - Moran Yang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Yiying Wang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Haiou Liu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China.
| | - Congjian Xu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, No. 128, Shenyang Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
122
|
Taguchi A, Furusawa A, Ito K, Nakajima Y, Shimizuguchi T, Hara K, Takao M, Kashiyama T, Kino N, Karasawa K, Yasugi T. Postradiotherapy persistent lymphopenia as a poor prognostic factor in patients with cervical cancer receiving radiotherapy: a single-center, retrospective study. Int J Clin Oncol 2020; 25:955-962. [PMID: 31960184 DOI: 10.1007/s10147-020-01623-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/06/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Radiotherapy (RT) is effective in cervical cancer; radiation-induced lymphopenia correlates with poor survival outcome in several cancer types. We investigated the association of total lymphocyte count (TLC) with survival outcomes in patients with cervical cancer. METHODS We retrospectively reviewed 168 patients with cervical cancer initially treated with definitive RT. We obtained clinicopathological data and TLCs before RT and at the end and at 6 months after RT. Patient-, treatment-, and tumor-specific factors were evaluated to determine their predictive values for overall survival. The association of overall and progression-free survivals with lymphopenia at each point was evaluated. RESULTS Median follow-up duration was 44 (interquartile range: 25-67) months. Median TLCs before RT and at the end and at 6 months after RT were 1625/mm3, 400/mm3, and 800/mm3 (interquartile range: 1270-1930/mm3, 290-550/mm3, and 600-1067/mm3), respectively. For overall survival, in addition to FIGO stage, body mass index, histology, treatment, and presence of para-aortic lymph node metastasis, lymphopenia at 6 months after RT was a poor prognostic factor in multivariate analysis (P = 0.0026; hazard ratio [HR], 3.06; 95% confidence interval [CI]: 1.48-6.33). For progression-free survival, TLCs before and at 6 months after RT were poor prognostic factors in univariate analysis (P = 0.0318 and 0.0081, respectively); however, the latter was the only independent prognostic factor in multivariate analysis (P = 0.0021; HR, 2.67; 95% CI: 1.43-4.99). CONCLUSION Post-RT persistent lymphopenia could be a poor prognostic factor for patients with cervical cancer who receive RT.
Collapse
Affiliation(s)
- Ayumi Taguchi
- Department of Gynecology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan.,Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Akiko Furusawa
- Department of Gynecology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan.
| | - Kei Ito
- Division of Radiation Oncology, Department of Radiology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Yujiro Nakajima
- Division of Radiation Oncology, Department of Radiology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan.,Department of Radiation Oncology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Takuya Shimizuguchi
- Division of Radiation Oncology, Department of Radiology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Konan Hara
- Graduate School of Economics, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-00333, Japan.,Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Maki Takao
- Department of Gynecology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Tomoko Kashiyama
- Department of Gynecology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan.,Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Nao Kino
- Department of Gynecology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Katsuyuki Karasawa
- Division of Radiation Oncology, Department of Radiology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Toshiharu Yasugi
- Department of Gynecology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| |
Collapse
|
123
|
Abstract
Checkpoint inhibitor therapy (CIT) has revolutionized cancer treatment but it has also reached a standstill when an absent dialog between cancer and immune cells makes it irrelevant. This occurs with high prevalence in the context of "immune silent" and, even perhaps, "immune-excluded" tumors. The latter are characterized by T cells restricted to the periphery of cancer nests. Since in either case T cells do not come in direct contact with most cancer cells, CIT rests immaterial. Adoptive cell therapy (ACT), may also be affected by limited access to antigen-bearing cancer cells. While lack of immunogenicity intuitively explains the immune silent phenotype, immune exclusion is perplexing. The presence of T cells at the periphery suggests that chemo-attraction recruits them and an immunogenic stimulus promotes their persistence. However, what stops the T cells from infiltrating the tumors' nests and reaching the germinal center (GC)? Possibly, a concentric gradient of increased chemo-repulsion or decreased chemo-attraction demarcates an abrupt "do not trespass" warning. Various hypotheses suggest physical or functional barriers but no definitive consensus exists over the weight that each plays in human cancers. On one hand, it could be hypothesized that the intrinsic biology of cancer cells may degenerate from a "cancer stem cell" (CSC)-like phenotype in the GC toward a progressively more immunogenic phenotype prone to immunogenic cell death (ICD) at the periphery. On the other hand, the intrinsic biology of the cancer cells may not change but it is the disorderly architecture of the tumor microenvironment (TME) that alters in a centripetal direction cancer cell metabolism, both directly and indirectly, the function of surrounding stromal cells. In this chapter, we examine whether the paradoxical exclusion of T cells from tumors may serve as a model to understand the requirements for tumor immune infiltration and, correspondingly, we put forth strategies to restore the dialog between immune cells and cancer to enhance the effectiveness of immune oncology (IO) approaches.
Collapse
Affiliation(s)
- Sara I Pai
- Massachusetts General Hospital, Harvard University, Boston, MA, USA.
| | | | | |
Collapse
|
124
|
Lodyga M, Hinz B. TGF-β1 - A truly transforming growth factor in fibrosis and immunity. Semin Cell Dev Biol 2019; 101:123-139. [PMID: 31879265 DOI: 10.1016/j.semcdb.2019.12.010] [Citation(s) in RCA: 267] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 12/20/2022]
Abstract
'Jack of all trades, master of everything' is a fair label for transforming growth factor β1 (TGF-β) - a cytokine that controls our life at many levels. In the adult organism, TGF-β1 is critical for the development and maturation of immune cells, maintains immune tolerance and homeostasis, and regulates various aspects of immune responses. Following acute tissue damages, TGF-β1 becomes a master regulator of the healing process with impacts on about every cell type involved. Divergence from the tight control of TGF-β1 actions, for instance caused by chronic injury, severe trauma, or infection can tip the balance from regulated physiological to excessive pathological repair. This condition of fibrosis is characterized by accumulation and stiffening of collagenous scar tissue which impairs organ functions to the point of failure. Fibrosis and dysregulated immune responses are also a feature of cancer, in which tumor cells escape immune control partly by manipulating TGF-β1 regulation and where immune cells are excluded from the tumor by fibrotic matrix created during the stroma 'healing' response. Despite the obvious potential of TGF-β-signalling therapies, globally targeting TGF-β1 receptor, downstream pathways, or the active growth factor have proven to be extremely difficult if not impossible in systemic treatment regimes. However, TGF-β1 binding to cell receptors requires prior activation from latent complexes that are extracellularly presented on the surface of immune cells or within the extracellular matrix. These different locations have led to some divergence in the field which is often either seen from the perspective of an immunologists or a fibrosis/matrix researcher. Despite these human boundaries, there is considerable overlap between immune and tissue repair cells with respect to latent TGF-β1 presentation and activation. Moreover, the mechanisms and proteins employed by different cells and spatiotemporal control of latent TGF-β1 activation provide specificity that is amenable to drug development. This review aims at synthesizing the knowledge on TGF-β1 extracellular activation in the immune system and in fibrosis to further stimulate cross talk between the two research communities in solving the TGF-β conundrum.
Collapse
Affiliation(s)
- Monika Lodyga
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, M5G1G6, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, M5G1G6, Canada.
| |
Collapse
|
125
|
Elkoshi Z. The Binary Classification Of Chronic Diseases. J Inflamm Res 2019; 12:319-333. [PMID: 31908517 PMCID: PMC6927256 DOI: 10.2147/jir.s227279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/07/2019] [Indexed: 12/19/2022] Open
Abstract
Acute diseases start with an insult and end when insult disappears. If the trauma induces an immune reaction (which happens in most cases), this reaction must be terminated with some type of resolution mechanism, when the cause of the trauma ceases. Chronicity develops if insult is permanent or if the resolution mechanism is defective. Another way to reach disease chronicity is a positive feedback loop, whereby the immune reaction activates an internal, insult-like reaction. A distinction between chronic states characterized by a persistent, low suppressive effect and those characterized by a persistent, high suppressive effect of regulatory T cells (Treg), is proposed. This two-class division represents two ways to reach chronicity: (a) by maintaining inflammatory reaction long after insult disappears ("low Treg"), or (b) by suppressing inflammatory reaction prior to the disappearance of insult ("high Treg"). This two-class division may explain the strong association between certain pathogens and cancer, on one hand, and between several other pathogens and autoimmunity, on the other hand. The weak association between autoimmune diseases and HIV infection and the relatively weak association between autoimmune diseases and cancer may be elucidated as well. In addition, the model rationalizes why immune-modulating drugs, which are effective in cancer, are also effective in "high Treg" viral infections, while corticosteroids, which are generally effective in autoimmune diseases, are also effective in other "low Treg" diseases (such as asthma, atopic dermatitis, and "low Treg" infections) but are not effective in solid malignancies and "high Treg" infections. Moreover, the model expounds why certain bacteria inhibit tumor growth and why these very bacteria induce autoimmune diseases.
Collapse
Affiliation(s)
- Zeev Elkoshi
- Taro Pharmaceutical Industries, Haifa Bay, Israel
| |
Collapse
|
126
|
Park J, Choi Y, Chang H, Um W, Ryu JH, Kwon IC. Alliance with EPR Effect: Combined Strategies to Improve the EPR Effect in the Tumor Microenvironment. Theranostics 2019; 9:8073-8090. [PMID: 31754382 PMCID: PMC6857053 DOI: 10.7150/thno.37198] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 09/16/2019] [Indexed: 12/16/2022] Open
Abstract
The use of nanomedicine for cancer treatment takes advantage of its preferential accumulation in tumors owing to the enhanced permeability and retention (EPR) effect. The development of cancer nanomedicine has promised highly effective treatment options unprecedented by standard therapeutics. However, the therapeutic efficacy of passively targeted nanomedicine is not always satisfactory because it is largely influenced by the heterogeneity of the intensity of the EPR effect exhibited within a tumor, at different stages of a tumor, and among individual tumors. In addition, limited data on EPR effectiveness in human hinders further clinical translation of nanomedicine. This unsatisfactory therapeutic outcome in mice and humans necessitates novel approaches to improve the EPR effect. This review focuses on current attempts at overcoming the limitations of traditional EPR-dependent nanomedicine by incorporating supplementary strategies, such as additional molecular targeting, physical alteration, or physiological remodeling of the tumor microenvironment. This review will provide valuable insight to researchers who seek to overcome the limitations of relying on the EPR effect alone in cancer nanomedicine and go "beyond the EPR effect".
Collapse
Affiliation(s)
- Jooho Park
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Yongwhan Choi
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Hyeyoun Chang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
| | - Wooram Um
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Ju Hee Ryu
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Ick Chan Kwon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
- Department of Cancer Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, United States
| |
Collapse
|
127
|
Istomine R, Alvarez F, Almadani Y, Philip A, Piccirillo CA. The Deubiquitinating Enzyme Ubiquitin-Specific Peptidase 11 Potentiates TGF-β Signaling in CD4 + T Cells to Facilitate Foxp3 + Regulatory T and T H17 Cell Differentiation. THE JOURNAL OF IMMUNOLOGY 2019; 203:2388-2400. [PMID: 31554694 DOI: 10.4049/jimmunol.1801689] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 08/26/2019] [Indexed: 12/16/2022]
Abstract
Foxp3+ regulatory T (TREG) cells are central mediators in the control of peripheral immune responses. Genome-wide transcriptional profiles show canonical signatures for Foxp3+ TREG cells, distinguishing them from Foxp3- effector T (TEFF) cells. We previously uncovered distinct mRNA translational signatures differentiating CD4+ TEFF and TREG cells through parallel measurements of cytosolic (global) and polysome-associated (translationally enhanced) mRNA levels in both subsets. We show that the mRNA encoding for the ubiquitin-specific peptidase 11 (USP11), a known modulator of TGF-β signaling, was preferentially translated in TCR-activated TREG cells compared with conventional, murine CD4+ T cells. TGF-β is a key cytokine driving the induction and maintenance of Foxp3 expression in T cells. We hypothesized that differential translation of USP11 mRNA endows TREG cells with an advantage to respond to TGF-β signals. In an in vivo mouse model promoting TREG cells plasticity, we found that USP11 protein was expressed at elevated levels in stable TREG cells, whereas ectopic USP11 expression enhanced the suppressive capacity and lineage commitment of these cells in vitro and in vivo. USP11 overexpression in TEFF cells enhanced the activation of the TGF-β pathway and promoted TREG or TH17, but not Th1, cell differentiation in vitro and in vivo, an effect abrogated by USP11 gene silencing or the inhibition of enzymatic activity. Thus, USP11 potentiates TGF-β signaling in both TREG and TEFF cells, in turn driving increased suppressive function and lineage commitment in thymic-derived TREG cells and potentiating the TGF-β-dependent differentiation of TEFF cells to peripherally induced TREG and TH17 cells.
Collapse
Affiliation(s)
- Roman Istomine
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada.,Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada.,Centre of Excellence in Translational Immunology, Montreal, Quebec H4A 3J1, Canada; and
| | - Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada.,Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada.,Centre of Excellence in Translational Immunology, Montreal, Quebec H4A 3J1, Canada; and
| | - Yasser Almadani
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec H3G 1A4, Canada; and.,Plastic Surgery Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Quebec H3G 1A4, Canada
| | - Anie Philip
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec H3G 1A4, Canada; and.,Plastic Surgery Research Laboratory, Research Institute of the McGill University Health Centre, Montreal, Quebec H3G 1A4, Canada
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada; .,Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada.,Centre of Excellence in Translational Immunology, Montreal, Quebec H4A 3J1, Canada; and
| |
Collapse
|
128
|
Rojas-Sanchez G, Cotzomi-Ortega I, Pazos-Salazar NG, Reyes-Leyva J, Maycotte P. Autophagy and Its Relationship to Epithelial to Mesenchymal Transition: When Autophagy Inhibition for Cancer Therapy Turns Counterproductive. BIOLOGY 2019; 8:biology8040071. [PMID: 31554173 PMCID: PMC6956138 DOI: 10.3390/biology8040071] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/14/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023]
Abstract
The manipulation of autophagy for cancer therapy has gained recent interest in clinical settings. Although inhibition of autophagy is currently being used in clinical trials for the treatment of several malignancies, autophagy has been shown to have diverse implications for normal cell homeostasis, cancer cell survival, and signaling to cells in the tumor microenvironment. Among these implications and of relevance for cancer therapy, the autophagic process is known to be involved in the regulation of protein secretion, in tumor cell immunogenicity, and in the regulation of epithelial-to-mesenchymal transition (EMT), a critical step in the process of cancer cell invasion. In this work, we have reviewed recent evidence linking autophagy to the regulation of EMT in cancer and normal epithelial cells, and have discussed important implications for the manipulation of autophagy during cancer therapy.
Collapse
Affiliation(s)
- Guadalupe Rojas-Sanchez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Israel Cotzomi-Ortega
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Nidia G Pazos-Salazar
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Puebla 72570, Mexico.
| | - Julio Reyes-Leyva
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Km 4.5 Carretera Atlixco-Metepec HGZ5, Puebla 74360, Mexico.
| | - Paola Maycotte
- Consejo Nacional de Ciencia y Tecnología (CONACYT)-CIBIOR, IMSS, Puebla 74360, Mexico.
| |
Collapse
|
129
|
Huang SH, McCann CD, Mota TM, Wang C, Lipkin SM, Jones RB. Have Cells Harboring the HIV Reservoir Been Immunoedited? Front Immunol 2019; 10:1842. [PMID: 31447850 PMCID: PMC6691121 DOI: 10.3389/fimmu.2019.01842] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/22/2019] [Indexed: 01/05/2023] Open
Abstract
Immunoediting is an important concept in oncology, delineating the mechanisms through which tumors are selected for resistance to immune-mediated elimination. The recent emergence of immunotherapies, such as checkpoint inhibitors, as pillars of cancer therapy has intensified interest in immunoediting as a constraint limiting the efficacy of these approaches. Immunoediting manifests at a number of levels for different cancers, for example through the establishment of immunosuppressive microenvironments within solid tumors. Of particular interest to the current review, selection also occurs at the cellular level; and recent studies have revealed novel mechanisms by which tumor cells acquire intrinsic resistance to immune recognition and elimination. While the selection of escape mutations in viral epitopes by HIV-specific T cells, which is a hallmark of chronic HIV infection, can be considered a form of immunoediting, few studies have considered the possibility that HIV-infected cells themselves may parallel tumors in having differential intrinsic susceptibilities to immune-mediated elimination. Such selection, on the level of an infected cell, may not play a significant role in untreated HIV, where infection is propagated by high levels of cell-free virus produced by cells that quickly succumb to viral cytopathicity. However, it may play an unappreciated role in individuals treated with effective antiretroviral therapy where viral replication is abrogated. In this context, an "HIV reservoir" persists, comprising long-lived infected cells which undergo extensive and dynamic clonal expansion. The ability of these cells to persist in infected individuals has generally been attributed to viral latency, thought to render them invisible to immune recognition, and/or to their compartmentalization in anatomical sites that are poorly accessible to immune effectors. Recent data from ex vivo studies have led us to propose that reservoir-harboring cells may additionally have been selected for intrinsic resistance to CD8+ T cells, limiting their elimination even in the context of antigen expression. Here, we draw on knowledge from tumor immunoediting to discuss potential mechanisms by which clones of HIV reservoir-harboring cells may resist elimination by CD8+ T cells. The establishment of such parallels may provide a premise for testing therapeutics designed to sensitize tumor cells to immune-mediated elimination as novel approaches aimed at curing HIV infection.
Collapse
Affiliation(s)
- Szu-Han Huang
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Chase D. McCann
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
- Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States
| | - Talia M. Mota
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Chao Wang
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Steven M. Lipkin
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - R. Brad Jones
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
- Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States
| |
Collapse
|
130
|
Braun E, Sauter D. Furin-mediated protein processing in infectious diseases and cancer. Clin Transl Immunology 2019; 8:e1073. [PMID: 31406574 PMCID: PMC6682551 DOI: 10.1002/cti2.1073] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/10/2019] [Accepted: 07/13/2019] [Indexed: 12/17/2022] Open
Abstract
Proteolytic cleavage regulates numerous processes in health and disease. One key player is the ubiquitously expressed serine protease furin, which cleaves a plethora of proteins at polybasic recognition motifs. Mammalian substrates of furin include cytokines, hormones, growth factors and receptors. Thus, it is not surprising that aberrant furin activity is associated with a variety of disorders including cancer. Furthermore, the enzymatic activity of furin is exploited by numerous viral and bacterial pathogens, thereby enhancing their virulence and spread. In this review, we describe the physiological and pathophysiological substrates of furin and discuss how dysregulation of a simple proteolytic cleavage event may promote infectious diseases and cancer. One major focus is the role of furin in viral glycoprotein maturation and pathogenicity. We also outline cellular mechanisms regulating the expression and activation of furin and summarise current approaches that target this protease for therapeutic intervention.
Collapse
Affiliation(s)
- Elisabeth Braun
- Institute of Molecular VirologyUlm University Medical CenterUlmGermany
| | - Daniel Sauter
- Institute of Molecular VirologyUlm University Medical CenterUlmGermany
| |
Collapse
|
131
|
Riddiough GE, Fifis T, Muralidharan V, Perini MV, Christophi C. Searching for the link; mechanisms underlying liver regeneration and recurrence of colorectal liver metastasis post partial hepatectomy. J Gastroenterol Hepatol 2019; 34:1276-1286. [PMID: 30828863 DOI: 10.1111/jgh.14644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/23/2019] [Accepted: 02/28/2019] [Indexed: 12/13/2022]
Abstract
Despite excellent treatment of primary colorectal cancer, the majority of deaths occur as a result of metastasis to the liver. Recent population studies have estimated that one quarter of patients with colorectal cancer will incur synchronous or metachronous colorectal liver metastasis. However, only one quarter of these patients will be eligible for potentially curative resection. Tumor recurrence occurs in reportedly 60% of patients undergoing hepatic resection, and the majority of intrahepatic recurrence occurs within the first 6 months of surgery. The livers innate ability to restore its homeostatic size, and volume facilitates major hepatic resection that currently offers the only chance of cure to patients with extensive hepatic metastases. Experimental and clinical evidence supports the notion that following partial hepatectomy, liver regeneration (LR) paradoxically drives tumor progression and increases the risk of recurrence. It is becoming increasingly clear that the processes that drive liver organogenesis, regeneration, and tumor progression are inextricably linked. This presents a major hurdle in the management of colorectal liver metastasis and other hepatic malignancies because therapies that reduce the risk of recurrence without hampering LR are sought. The processes and pathways underlying these phenomena are multiple, complex, and cross-communicate. In this review, we will summarize the common mechanisms contributing to both LR and tumor recurrence.
Collapse
Affiliation(s)
- Georgina E Riddiough
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Theodora Fifis
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | | | - Marcos V Perini
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Christopher Christophi
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
132
|
Dahmani A, Janelle V, Carli C, Richaud M, Lamarche C, Khalili M, Goupil M, Bezverbnaya K, Bramson JL, Delisle JS. TGFβ Programs Central Memory Differentiation in Ex Vivo-Stimulated Human T Cells. Cancer Immunol Res 2019; 7:1426-1439. [PMID: 31308016 DOI: 10.1158/2326-6066.cir-18-0691] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 03/27/2019] [Accepted: 07/09/2019] [Indexed: 11/16/2022]
Abstract
The adoptive transfer of ex vivo-expanded T cells is a promising approach to treat several malignancies. Several lines of evidence support that the infusion of T cells with early memory features, capable of expanding and persisting after transfer, are associated with better outcomes. We report herein that exposure to exogenous TGFβ during human T-cell stimulation ex vivo leads to the accumulation of early/central memory (Tcm) cells. Exposure to TGFβ suppressed the expression of BLIMP-1, a key orchestrator of effector T-cell differentiation, and led to the upregulation of the memory-associated transcription factor ID3. Accordingly, this was associated with an early memory transcriptional signature in both CD4+ and CD8+ T-cell subsets. The T cells stimulated in the presence of TGFβ expanded normally, and displayed polyfunctional features and no suppressive activity. The adoptive transfer of ex vivo-stimulated T cells into immunodeficient mice confirmed that TGFβ-conditioned cells had an enhanced capacity to persist and mediate xenogeneic graft-versus-host disease, as predicted by their early T-cell memory phenotype. Chimeric antigen receptor-expressing T cells generated in the presence of exogenous TGFβ were cytotoxic and more effective at controlling tumor growth in immunodeficient animals. This work unveils a new role for TGFβ in memory T-cell differentiation and indicates that TGFβ signaling may be harnessed to program Tcm differentiation in the context of ex vivo T-cell stimulation for adoptive immunotherapy in humans.
Collapse
Affiliation(s)
- Amina Dahmani
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Valérie Janelle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Cédric Carli
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Manon Richaud
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Caroline Lamarche
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Myriam Khalili
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Mathieu Goupil
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada
| | - Ksenia Bezverbnaya
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan L Bramson
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jean-Sébastien Delisle
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont (CRHMR), Montreal, Quebec, Canada.
- Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
- Hematology-Oncology Division, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| |
Collapse
|
133
|
Hu Q, Myers M, Fang W, Yao M, Brummer G, Hawj J, Smart C, Berkland C, Cheng N. Role of ALDH1A1 and HTRA2 expression in CCL2/CCR2-mediated breast cancer cell growth and invasion. Biol Open 2019; 8:bio.040873. [PMID: 31208996 PMCID: PMC6679398 DOI: 10.1242/bio.040873] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chemokines mediate immune cell trafficking during tissue development, wound healing and infection. The chemokine CCL2 is best known to regulate macrophage recruitment during wound healing, infection and inflammatory diseases. While the importance of CCL2/CCR2 signaling in macrophages during cancer progression is well documented, we recently showed that CCL2-mediated breast cancer progression depends on CCR2 expression in carcinoma cells. Using 3D Matrigel: Collagen cultures of SUM225 and DCIS.com breast cancer cells, this study characterized the mechanisms of CCL2/CCR2 signaling in cell growth and invasion. SUM225 cells, which expressed lower levels of CCR2 than DCIS.com cells, formed symmetrical spheroids in Matrigel: Collagen, and were not responsive to CCL2 treatment. DCIS.com cells formed asymmetric cell clusters in Matrigel: Collagen. CCL2 treatment increased growth, decreased expression of E-cadherin and increased TWIST1 expression. CCR2 overexpression in SUM225 cells increased responsiveness to CCL2 treatment, enhancing growth and invasion. These phenotypes corresponded to increased expression of Aldehyde Dehydrogenase 1A1 (ALDH1A1) and decreased expression of the mitochondrial serine protease HTRA2. CCR2 deficiency in DCIS.com cells inhibited CCL2-mediated growth and invasion, corresponding to decreased ALDH1A1 expression and increased HTRA2 expression. ALDH1A1 and HTRA2 expression were modulated in CCR2-deficient and CCR2-overexpressing cell lines. We found that ALDH1A1 and HTRA2 regulates CCR2-mediated breast cancer cell growth and cellular invasion in a CCL2/CCR2 context-dependent manner. These data provide novel insight on the mechanisms of chemokine signaling in breast cancer cell growth and invasion, with important implications on targeted therapeutics for anti-cancer treatment. This article has an associated First Person interview with the first author of the paper. Summary: Chemokines are known to regulate immune cell recruitment during inflammation. This report characterizes novel molecular mechanisms through which CCL2/CCR2 chemokine signaling in breast cancer cells regulates growth and invasion.
Collapse
Affiliation(s)
- Qingting Hu
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Megan Myers
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wei Fang
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Min Yao
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Gage Brummer
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Justin Hawj
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Curtis Smart
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Nikki Cheng
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA .,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
134
|
Ungefroren H. Blockade of TGF-β signaling: a potential target for cancer immunotherapy? Expert Opin Ther Targets 2019; 23:679-693. [PMID: 31232607 DOI: 10.1080/14728222.2019.1636034] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Malignant tumors often escape surveillance and eventual destruction by the host immune system through a variety of strategies including production of transforming growth factor (TGF)-β. Because of its generally immunosuppressive role, TGF-β has emerged as a promising therapeutic target in cancer immunotherapy. Areas covered: This article looks at specific mechanisms of how TGF-β controls the function of various immune cell subsets in the tumor microenvironment and focusses on T-cells. Various inhibition tools of TGF-β signaling and potential targets of therapeutic intervention are assessed along with the recent progress in combining TGF-β blockade and immune-mediated therapies. To round off the article, a summary of results from clinical trials is provided in which TGF-β blockade has shown therapeutic benefit for patients. Expert opinion: Data from preclinical models have shown that blocking TGF-β signaling can overcome resistance mechanisms and in combination with immune-checkpoint therapies, can yield additive or synergistic anti-tumor responses. The future of immunooncology will therefore be based on combination trials. Since response rates may critically depend on both cancer type and stage, selection of only those patients who can benefit from combinatorial immunotherapy regimens is of utmost importance.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- a First Department of Medicine , University Hospital Schleswig-Holstein, Campus Lübeck, and University of Lübeck , Lübeck , Germany.,b Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery , University Hospital Schleswig-Holstein , Campus Kiel, Kiel , Germany
| |
Collapse
|
135
|
Gupta A, Budhu S, Merghoub T. One checkpoint may hide another: inhibiting the TGFβ signaling pathway enhances immune checkpoint blockade. Hepatobiliary Surg Nutr 2019; 8:289-294. [PMID: 31245417 PMCID: PMC6561888 DOI: 10.21037/hbsn.2019.01.10] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/06/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Aditi Gupta
- Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy at Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sadna Budhu
- Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy at Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Immunology Program, Parker Institute for Cancer Immunotherapy at Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
136
|
Ungefroren H. TGF-β Signaling in Cancer: Control by Negative Regulators and Crosstalk with Proinflammatory and Fibrogenic Pathways. Cancers (Basel) 2019; 11:cancers11030384. [PMID: 30893848 PMCID: PMC6468838 DOI: 10.3390/cancers11030384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 11/16/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family of secreted growth factors controls many aspects of cell and tissue physiology in multicellular eukaryotes. Dysregulation of its pathway contributes to a broad variety of pathologies, including fibrosis and cancer. TGF-β acts as a powerful tumor suppressor in epithelial cells but during later stages of tumor development cancer cells eventually respond to this cytokine with epithelial-mesenchymal transition (EMT), invasion, metastasis, and immunosuppression. This collection of articles covers some important aspects of TGF-β signaling in cancer. Two articles focus on the role of TGF-β in tumor immunity and pro- and anti-inflammatory signaling, with one analyzing its impact on T-cell biology and different T-cell subsets, while the other deals with modulation of anti-inflammatory signaling by TGF-β receptors through proinflammatory signaling by immune receptors and the role of mechanotransduction in TGF-β-dependent immunosuppression. Another set of four chapters highlights the fact that context-dependent responsiveness to TGF-β is largely controlled by inputs from negative regulators and cooperation with proinflammatory and proapoptotic pathways. This theme is extended to the regulation of Smad signaling by differential phosphorylation, eventually converting canonical Smad signaling to a mitogenic, fibrogenic and carcinogenic outcome. Last, it is discussed how another posttranslational modification, SUMOylation, can modify protein function and impact TGF-β-induced EMT, invasion and metastasis.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany.
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany.
| |
Collapse
|
137
|
Prunk M, Nanut MP, Sabotic J, Svajger U, Kos J. Increased cystatin F levels correlate with decreased cytotoxicity of cytotoxic T cells. Radiol Oncol 2019; 53:57-68. [PMID: 30840596 PMCID: PMC6411024 DOI: 10.2478/raon-2019-0007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 01/05/2019] [Indexed: 12/13/2022] Open
Abstract
Background Cystatin F is a protein inhibitor of cysteine peptidases, expressed predominantly in immune cells and localised in endosomal/lysosomal compartments. In cytotoxic immune cells cystatin F inhibits both the major pro-granzyme convertases, cathepsins C and H that activate granzymes, and cathepsin L, that acts as perforin activator. Since perforin and granzymes are crucial molecules for target cell killing by cytotoxic lymphocytes, defects in the activation of either granzymes or perforin can affect their cytotoxic potential. Materials and methods Levels of cystatin F were assessed by western blot and interactions of cystatin F with cathepsins C, H and L were analysed by immunoprecipitation and confocal microscopy. In TALL-104 cells specific activities of the cathepsins and granzyme B were determined using peptide substrates. Results Two models of reduced T cell cytotoxicity of TALL-104 cell line were established, either by treatment by ionomycin or by immunosuppressive transforming growth factor beta. Reduced cytotoxicity correlated with increased levels of cystatin F and with attenuated activities of cathepsins C, H and L and of granzyme B. Co-localisation of cystatin F and cathepsins C, H and L and interactions between cystatin F and cathepsins C and H were demonstrated. Conclusions Cystatin F is designated as a possible regulator of T cell cytotoxicity, similar to its role in natural killer cells.
Collapse
Affiliation(s)
- Mateja Prunk
- Jožef Stefan Institute, Department of Biotechnology, Ljubljana, Slovenia
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | | | - Jerica Sabotic
- Jožef Stefan Institute, Department of Biotechnology, Ljubljana, Slovenia
| | - Urban Svajger
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Janko Kos
- Jožef Stefan Institute, Department of Biotechnology, Ljubljana, Slovenia
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
- Prof. Janko Kos, Ph.D., Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia.
Phone: +386 1 4769 604; Fax: +386 1 4258 031
| |
Collapse
|
138
|
Wu XZ, Zhai K, Yi FS, Wang Z, Wang W, Wang Y, Pei XB, Shi XY, Xu LL, Shi HZ. IL-10 promotes malignant pleural effusion in mice by regulating T H 1- and T H 17-cell differentiation and migration. Eur J Immunol 2019; 49:653-665. [PMID: 30695099 DOI: 10.1002/eji.201847685] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 12/10/2018] [Accepted: 01/28/2019] [Indexed: 01/23/2023]
Abstract
The role of IL-10 in malignant pleural effusion (MPE) remains unknown. By using murine MPE models, we observed that an increase in pleural IL-10 was a significant predictor of increased risk of death. We noted that TH 1- and TH 17-cell content in MPE was higher in IL-10-/- mice than in WT mice, and IL-10 deficiency promoted differentiation into TH 1 but not into TH 17 cells. A higher fraction of TH 1 and TH 17 cells in the MPE of IL-10-/- mice expressed CXCR3 compared with WT mice. We also demonstrated that Lewis lung cancer and colon adenocarcinoma cells secreted large amounts of CXCL10, a ligand of CXCR3, which induced the migration of TH 1 and TH 17 cells into the MPE, and IFN-γ could promote this signaling cascade. Furthermore, intrapleural injection of mice with CXCL10-deficient tumor cells led to decreased TH 1- and TH 17-cell content in MPE, increased MPE volume, and reduced survival of MPE-bearing mice. Taken together, we demonstrated that IL-10 deficiency promoted T-cell differentiation into TH 1 cells and upregulated the CXCR3-CXCL10 signaling pathway that recruits TH 1 and TH 17 cells into MPE, ultimately resulting in decreased MPE formation and longer survival time of mice-bearing MPE.
Collapse
Affiliation(s)
- Xiu-Zhi Wu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Kan Zhai
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Feng-Shuang Yi
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhen Wang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Wen Wang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yao Wang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xue-Bin Pei
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xin-Yu Shi
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Li-Li Xu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Huan-Zhong Shi
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
139
|
TGFβ Imprinting During Activation Promotes Natural Killer Cell Cytokine Hypersecretion. Cancers (Basel) 2018; 10:cancers10110423. [PMID: 30400618 PMCID: PMC6267005 DOI: 10.3390/cancers10110423] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 10/31/2018] [Indexed: 12/18/2022] Open
Abstract
Transforming growth factor-beta (TGFβ) is a potent immunosuppressive cytokine that inhibits the anti-tumor responses of NK cells and T cells. However, the stimulation of natural killer (NK) cells with pro-inflammatory cytokines decreases NK cell sensitivity to TGFβ. Herein, we sought to determine if TGFβ imprinting (TGFβi) during NK cell activation and expansion would decrease NK cell sensitivity to TGFβ suppression. To this end, we demonstrate that the activation of NK cells during chronic IL-2 stimulation and TGFβi potently induces NK cell hypersecretion of interferon-gamma (IFNγ) and tumor necrosis factor-alpha (TNFα) in response to tumor targets which persists for at least one month in vitro after the removal of TGFβ. TGFβi NK cell cytokine hypersecretion is induced following both cytokine and tumor activation. Further, TGFβi NK cells have a marked suppression of SMAD3 and T-bet which is associated with altered chromatin accessibility. In contrast to their heightened cytokine secretion, TGFβi NK cells downregulate several activating receptors, granzyme and perforin, and upregulate TRAIL, leading to cell-line-specific alterations in cytotoxicity. These findings may impact our understanding of how TGFβ affects NK cell development and anti-tumor function.
Collapse
|