112 results on '"Kryczek I"'
Search Results
2. 23 INVITED T regs in cancer
- Author
-
curiel, T., primary, Barnett, B., additional, Zou, W., additional, Kryczek, I., additional, Brumlik, M., additional, Cheng, J., additional, and Rueter, J., additional
- Published
- 2006
- Full Text
- View/download PDF
3. A phase II trial of denileukin diftitox to treat refractory advanced-stage ovarian cancer
- Author
-
Barnett, B. G., primary, Ruter, J., additional, Brumlik, M. J., additional, Kryczek, I., additional, Cheng, P. J., additional, Zou, W., additional, and Curiel, T. J., additional
- Published
- 2006
- Full Text
- View/download PDF
4. Depleting Regulatory T Cells is Associated with Improved Immunity and Tumor Clearance in Human Cancer
- Author
-
Barnett, B., primary, Kryczek, I., additional, Cheng, P., additional, Zou, W., additional, and Curiel, T.J., additional
- Published
- 2005
- Full Text
- View/download PDF
5. 290 DEPLETING REGULATORY T CELLS IS ASSOCIATED WITH IMPROVED IMMUNITY AND TUMOR CLEARANCE IN HUMAN CANCER
- Author
-
Barnett, B., primary, Kryczek, I., additional, Cheng, P., additional, Zou, W., additional, and Curiel, T. J., additional
- Published
- 2005
- Full Text
- View/download PDF
6. IL-6 production in ovarian carcinoma is associated with histiotype and biological characteristics of the tumour and influences local immunity
- Author
-
Kryczek, I, primary, Gryboś, M, additional, Karabon, L, additional, Klimczak, A, additional, and Lange, A, additional
- Published
- 2000
- Full Text
- View/download PDF
7. CXCL12 and vascular endothelial growth factor synergistically induce neonaniogenisis in human ovarian cancers
- Author
-
Kryczek, I., Andrzej Lange, Mottram, P., Alvarez, X., Cheng, P., Hogan, M., Moons, L., Wei, S., Zou, L., Machelon, V., Emilie, D., Terrassa, M., Lackner, A., Curiel, T. J., Carmeliet, P., and Zou, W.
8. Endogenous interleukin-10 constrains Th17 cells in patients with inflammatory bowel disease
- Author
-
Wilke Cailin M, Wang Lin, Wei Shuang, Kryczek Ilona, Huang Emina, Kao John, Lin Yanwei, Fang Jingyuan, and Zou Weiping
- Subjects
Th17 ,IL-10 ,IL-1 ,IL-17 ,inflammation ,Crohn's disease ,Medicine - Abstract
Abstract Background Th17 cells play a role in inflammation. Interleukin (IL)-10 is a potent anti-inflammatory cytokine. However, it is poorly understood whether and how endogenous IL-10 impacts the development of Th17 cells in human pathologies. Materials and methods We examined the relationship between IL-10 and Th17 cells in patients with Crohn's disease and in IL-10-deficient (IL-10-/-) mice. Th17 cells and dendritic cells (DCs) were defined by flow cytometry and evaluated by functional studies. Results We detected elevated levels of IL-17 and Th17 cells in the intestinal mucosa of patients with Crohn's disease. Intestinal DCs from Crohn's patients produced more IL-1β than controls and were superior to blood DCs in Th17 induction through an IL-1-dependent mechanism. Furthermore, IL-17 levels were negatively associated with those of IL-10 and were positively associated those of IL-1β in intestinal mucosa. These data point toward an in vivo cellular and molecular link among endogenous IL-10, IL-1, and Th17 cells in patients with Crohn's disease. We further investigated this relationship in IL-10-/- mice. We observed a systemic increase in Th17 cells in IL-10-/- mice when compared to wild-type mice. Similar to the intestinal DCs in patients with Crohn's disease, murine IL-10-/- DCs produced more IL-1β than their wild-type counterparts and promoted Th17 cell development in an IL-1-dependent manner. Finally, in vivo blockade of IL-1 receptor signaling reduced Th17 cell accumulation and inflammation in a mouse model of chemically-induced colitis. Conclusions Endogenous IL-10 constrains Th17 cell development through the control of IL-1 production by DCs, and reaffirms the crucial anti-inflammatory role of IL-10 in patients with chronic inflammation.
- Published
- 2011
- Full Text
- View/download PDF
9. The UBA1-STUB1 axis mediates cancer immune escape and resistance to checkpoint blockade.
- Author
-
Bao Y, Cruz G, Zhang Y, Qiao Y, Mannan R, Hu J, Yang F, Gondal M, Shahine M, Kang S, Mahapatra S, Chu A, Choi JE, Yu J, Lin H, Miner SJ, Robinson DR, Wu YM, Zheng Y, Cao X, Su F, Wang R, Hosseini N, Cieslik M, Kryczek I, Vaishampayan U, Zou W, and Chinnaiyan AM
- Abstract
How cancer cells escape immune surveillance and resist immune checkpoint blockade (ICB) remains to be fully elucidated. By screening candidate genes frequently gained in cancer, we identified expression of ubiquitin-like modifier activating enzyme 1 (UBA1) as being the most negatively correlated with signatures related to effector CD8+ T-cells. High UBA1 expression was strongly predictive of treatment resistance and poor survival in ICB cohorts. Functional studies revealed that UBA1 mediated immune escape to promote tumor growth. Immune profiling further showed that Uba1 overexpression or depletion markedly decreased or increased functional intratumoral CD8+ T-cells, respectively. Importantly, a selective UBA1 inhibitor, TAK-243, significantly synergized with ICB in multiple syngeneic models. Mechanistically, depletion or inactivation of the UBA1-STUB1 axis stabilized a key interferon pathway component (JAK1), enhanced IFN-signaling, and elevated key immune modulators, including CXCL9, CXCL10, and MHC class I. Our study warrants clinical evaluation of the combination of UBA1 inhibitors and ICB.
- Published
- 2024
- Full Text
- View/download PDF
10. Phase II trial of multi-tyrosine kinase inhibitor ESK981 in combination with PD-1 inhibitor nivolumab in patients with metastatic castration-resistant prostate cancer.
- Author
-
Heath EI, Chen W, Choi JE, Dobson K, Smith M, Maj T, Kryczek I, Zou W, Chinnaiyan AM, and Qiao Y
- Abstract
Increasing the response rates of immune checkpoint inhibitors in patients with metastatic castration-resistant prostate cancer (mCRPC) presents a significant challenge. ESK981 is a multi-tyrosine kinase and PIKfyve lipid kinase inhibitor that augments immunotherapeutic responses. In this phase II study, ESK981 was combined with the PD-1 blocking monoclonal antibody nivolumab to test for potentially improved response rates in patients with mCRPC who have progressed on androgen receptor (AR)-targeted agents and chemotherapy. Eligible patients received ESK981 orally once daily for five consecutive days, followed by a two-day break. Patients were also treated with nivolumab intravenously on Day 1 of each 28-day cycle. The primary endpoints were a 50% reduction in prostate-specific antigen (PSA50), and safety. Secondary endpoints included radiographic progression free survival (rPFS) and overall survival (OS). Additional investigations included whole exome sequencing in patients. Ten patients were enrolled. The maximum PSA decline from baseline of 14% was achieved in only one patient. Grade 3 treatment-related adverse events (AEs) included fatigue, anemia, and lymphopenia. There were no Grade 4 events. The median rPFS was 3.7 months (95% CI, 1.6-8.4). The median OS was 9.6 months (95% CI, 1.8-22.4). The study was terminated due to futility after 10 patients. Whole exome sequencing identified AR amplification in 63% of patients (5/8). ESK981 + nivolumab showed no antitumor activity in patients with AR-positive (AR+) mCRPC. Further evaluation of ESK981 combined with the PD-1 inhibitor nivolumab in AR + mCRPC patients is not warranted. (Trial registration: ClinicalTrials.gov NCT04159896. Registration date: November 12, 2019.)., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
11. Itaconate transporter SLC13A3 impairs tumor immunity via endowing ferroptosis resistance.
- Author
-
Lin H, Tison K, Du Y, Kirchhoff P, Kim C, Wang W, Yang H, Pitter M, Yu J, Liao P, Zhou J, Vatan L, Grove S, Wei S, Vigil T, Shah YM, Mortensen R, Kryczek I, Garmire L, Sivaccumar JP, Ramesh AK, Zhang N, An Z, Wang S, and Zou W
- Abstract
Immune checkpoint blockade (ICB) triggers tumor ferroptosis. However, most patients are unresponsive to ICB. Tumors might evade ferroptosis in the tumor microenvironment (TME). Here, we discover SLC13A3 is an itaconate transporter in tumor cells and endows tumor ferroptosis resistance, diminishing tumor immunity and ICB efficacy. Mechanistically, tumor cells uptake itaconate via SLC13A3 from tumor-associated macrophages (TAMs), thereby activating the NRF2-SLC7A11 pathway and escaping from immune-mediated ferroptosis. Structural modeling and molecular docking analysis identify a functional inhibitor for SLC13A3 (SLC13A3i). Deletion of ACOD1 (an essential enzyme for itaconate synthesis) in macrophages, genetic ablation of SLC13A3 in tumors, or treatment with SLC13A3i sensitize tumors to ferroptosis, curb tumor progression, and bolster ICB effectiveness. Thus, we identify the interplay between tumors and TAMs via the SLC13A3-itaconate-NRF2-SLC7A11 axis as a previously unknown immune ferroptosis resistant mechanism in the TME and SLC13A3 as a promising immunometabolic target for treating SLC13A3
+ cancer., Competing Interests: Declaration of interests W.Z. serves as a scientific advisor or consultant for Cstone, Hanchor Bio, and NextCure. S. Wang is a consultant for Proteovant Therapeutics and owns stock in Roivant Sciences. S. Wang is a co-founder, paid consultant, and member of the board of directors of Ascentage Pharma Group International and owns stock in Ascentage. N.Z., Z.A., J.P.S., A.K.R., W.Z., H.L., S. Wang, and P.K. are listed inventors in a patent application (UTSH.P0407US.P1) related to this work., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
12. Phase II trial of multi-kinase inhibitor ESK981 in patients with metastatic castration-resistant prostate cancer.
- Author
-
Heath EI, Chen W, Heilbrun L, Choi JE, Dobson K, Smith M, Maj T, Vaishampayan U, Kryczek I, Zou W, Chinnaiyan AM, and Qiao Y
- Abstract
ESK981 is a potent tyrosine kinase and PIKfyve lipid kinase inhibitor. This phase II trial evaluated the efficacy of ESK981 as a single agent in patients with androgen receptor-positive (AR +) metastatic castration-resistant prostate cancer (mCRPC). Eligible patients had mCRPC with progression on AR-targeted agents and without prior chemotherapy treatment. Each patient received 160 mg ESK981 once daily for 5 days per week for 4 weeks per cycle (except for an adverse event (AE) occurrence). The primary endpoints were a 50% reduction in prostate-specific antigen (PSA50), and safety. Secondary endpoints included the time and the duration of PSA response, PSA progression rates, PSA progression free survival (PFS) and overall survival (OS). Exploratory investigations included whole exome sequencing in patients before treatment, and morphological evaluation of biopsy samples pre- and post-treatment. PSA was evaluated in 13 patients. Only one patient (7.7% two-sided 95% Wilson CI (0.4%, 33.3%)) experienced a reduction in their PSA levels by 50% or more. The most common grade 3 treatment-related AEs were cardiac disorders, diarrhea, hypertension, alanine transaminase and aspartate transaminase elevations. No grade 4-5 events occurred. Median PFS was 1.8 months, and median OS was 12.1 months. Peripheral immune cells showed increased T cell activation and cytokine production in two patients who received 12-weeks of ESK981. Although relatively well tolerated, ESK981 alone showed no anti-tumor activity in patients with AR + mCRPC and its further evaluation as a single agent in AR + mCRPC is not warranted. (Trial registration: ClinicalTrials.gov, NCT03456804. Registration date: March 7, 2018)., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
13. Progestogen-driven B7-H4 contributes to onco-fetal immune tolerance.
- Author
-
Yu J, Yan Y, Li S, Xu Y, Parolia A, Rizvi S, Wang W, Zhai Y, Xiao R, Li X, Liao P, Zhou J, Okla K, Lin H, Lin X, Grove S, Wei S, Vatan L, Hu J, Szumilo J, Kotarski J, Freeman ZT, Skala S, Wicha M, Cho KR, Chinnaiyan AM, Schon S, Wen F, Kryczek I, Wang S, Chen L, and Zou W
- Subjects
- Animals, Female, Humans, Mice, Pregnancy, Breast Neoplasms immunology, Breast Neoplasms genetics, Breast Neoplasms metabolism, Receptors, Progesterone metabolism, Transcription Factors metabolism, Cell Line, Tumor, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Mice, Inbred C57BL, Placenta metabolism, Placenta immunology, V-Set Domain-Containing T-Cell Activation Inhibitor 1 metabolism, Immune Tolerance, Progestins pharmacology, Progestins metabolism, Progesterone metabolism
- Abstract
Immune tolerance mechanisms are shared in cancer and pregnancy. Through cross-analyzing single-cell RNA-sequencing data from multiple human cancer types and the maternal-fetal interface, we found B7-H4 (VTCN1) is an onco-fetal immune tolerance checkpoint. We showed that genetic deficiency of B7-H4 resulted in immune activation and fetal resorption in allogeneic pregnancy models. Analogously, B7-H4 contributed to MPA/DMBA-induced breast cancer progression, accompanied by CD8
+ T cell exhaustion. Female hormone screening revealed that progesterone stimulated B7-H4 expression in placental and breast cancer cells. Mechanistically, progesterone receptor (PR) bound to a newly identified -58 kb enhancer, thereby mediating B7-H4 transcription via the PR-P300-BRD4 axis. PR antagonist or BRD4 degrader potentiated immunotherapy in a murine B7-H4+ breast cancer model. Thus, our work unravels a mechanistic and biological connection of a female sex hormone (progesterone) to onco-fetal immune tolerance via B7-H4 and suggests that the PR-P300-BRD4 axis is targetable for treating B7-H4+ cancer., Competing Interests: Declaration of interests W.Z. has served as a scientific advisor or consultant for Cstone, NextCure, and Hanchorbio. S. Wang is a co-founder and paid consultant of Ascentage Pharma Group International and owns stock in Ascentage. L.C. has been a scientific founder, consultant, and/or board observer for NextCure, Normunity, Tayu, Zai Lab, Tpioneer, Vcanbio, OncoC4, and GenomiCare and has sponsored research funds from NextCure, Normunity, and DynamiCure. This research is conducted independently and has not received resources from and is unrelated to the scientific and commercial pursuits of these industrial entities, including NextCure., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
14. PIKfyve controls dendritic cell function and tumor immunity.
- Author
-
Choi JE, Qiao Y, Kryczek I, Yu J, Gurkan J, Bao Y, Gondal M, Tien JC, Maj T, Yazdani S, Parolia A, Xia H, Zhou J, Wei S, Grove S, Vatan L, Lin H, Li G, Zheng Y, Zhang Y, Cao X, Su F, Wang R, He T, Cieslik M, Green MD, Zou W, and Chinnaiyan AM
- Abstract
The modern armamentarium for cancer treatment includes immunotherapy and targeted therapy, such as protein kinase inhibitors. However, the mechanisms that allow cancer-targeting drugs to effectively mobilize dendritic cells (DCs) and affect immunotherapy are poorly understood. Here, we report that among shared gene targets of clinically relevant protein kinase inhibitors, high PIKFYVE expression was least predictive of complete response in patients who received immune checkpoint blockade (ICB). In immune cells, high PIKFYVE expression in DCs was associated with worse response to ICB. Genetic and pharmacological studies demonstrated that PIKfyve ablation enhanced DC function via selectively altering the alternate/non-canonical NF-κB pathway. Both loss of Pikfyve in DCs and treatment with apilimod, a potent and specific PIKfyve inhibitor, restrained tumor growth, enhanced DC-dependent T cell immunity, and potentiated ICB efficacy in tumor-bearing mouse models. Furthermore, the combination of a vaccine adjuvant and apilimod reduced tumor progression in vivo . Thus, PIKfyve negatively controls DCs, and PIKfyve inhibition has promise for cancer immunotherapy and vaccine treatment strategies.
- Published
- 2024
- Full Text
- View/download PDF
15. PIKfyve, expressed by CD11c-positive cells, controls tumor immunity.
- Author
-
Choi JE, Qiao Y, Kryczek I, Yu J, Gurkan J, Bao Y, Gondal M, Tien JC, Maj T, Yazdani S, Parolia A, Xia H, Zhou J, Wei S, Grove S, Vatan L, Lin H, Li G, Zheng Y, Zhang Y, Cao X, Su F, Wang R, He T, Cieslik M, Green MD, Zou W, and Chinnaiyan AM
- Subjects
- Animals, Female, Humans, Mice, Cell Line, Tumor, Hydrazones, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Immunotherapy methods, Mice, Inbred C57BL, Neoplasms immunology, Neoplasms genetics, Neoplasms therapy, NF-kappa B metabolism, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Pyrimidines, T-Lymphocytes immunology, Male, CD11c Antigen metabolism, Dendritic Cells immunology, Dendritic Cells metabolism, Dendritic Cells drug effects, Morpholines pharmacology, Phosphatidylinositol 3-Kinases metabolism
- Abstract
Cancer treatment continues to shift from utilizing traditional therapies to targeted ones, such as protein kinase inhibitors and immunotherapy. Mobilizing dendritic cells (DC) and other myeloid cells with antigen presenting and cancer cell killing capacities is an attractive but not fully exploited approach. Here, we show that PIKFYVE is a shared gene target of clinically relevant protein kinase inhibitors and high expression of this gene in DCs is associated with poor patient response to immune checkpoint blockade (ICB) therapy. Genetic and pharmacological studies demonstrate that PIKfyve ablation enhances the function of CD11c
+ cells (predominantly dendritic cells) via selectively altering the non-canonical NF-κB pathway. Both loss of Pikfyve in CD11c+ cells and treatment with apilimod, a potent and specific PIKfyve inhibitor, restrained tumor growth, enhanced DC-dependent T cell immunity, and potentiated ICB efficacy in tumor-bearing mouse models. Furthermore, the combination of a vaccine adjuvant and apilimod reduced tumor progression in vivo. Thus, PIKfyve negatively regulates the function of CD11c+ cells, and PIKfyve inhibition has promise for cancer immunotherapy and vaccine treatment strategies., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
16. PAD4 controls tumor immunity via restraining the MHC class II machinery in macrophages.
- Author
-
Pitter MR, Kryczek I, Zhang H, Nagarsheth N, Xia H, Wu Z, Tian Y, Okla K, Liao P, Wang W, Zhou J, Li G, Lin H, Vatan L, Grove S, Wei S, Li Y, and Zou W
- Subjects
- Mice, Animals, Protein-Arginine Deiminases metabolism, Protein-Arginine Deiminase Type 4 genetics, Protein-Arginine Deiminase Type 4 metabolism, Histocompatibility Antigens Class II metabolism, Macrophages metabolism, Hydrolases metabolism, Protein Processing, Post-Translational
- Abstract
Tumor-associated macrophages (TAMs) shape tumor immunity and therapeutic efficacy. However, it is poorly understood whether and how post-translational modifications (PTMs) intrinsically affect the phenotype and function of TAMs. Here, we reveal that peptidylarginine deiminase 4 (PAD4) exhibits the highest expression among common PTM enzymes in TAMs and negatively correlates with the clinical response to immune checkpoint blockade. Genetic and pharmacological inhibition of PAD4 in macrophages prevents tumor progression in tumor-bearing mouse models, accompanied by an increase in macrophage major histocompatibility complex (MHC) class II expression and T cell effector function. Mechanistically, PAD4 citrullinates STAT1 at arginine 121, thereby promoting the interaction between STAT1 and protein inhibitor of activated STAT1 (PIAS1), and the loss of PAD4 abolishes this interaction, ablating the inhibitory role of PIAS1 in the expression of MHC class II machinery in macrophages and enhancing T cell activation. Thus, the PAD4-STAT1-PIAS1 axis is an immune restriction mechanism in macrophages and may serve as a cancer immunotherapy target., Competing Interests: Declaration of interests W.Z. has served as a scientific advisor or consultant for Cstone, NextCure, and HanchorBio, Inc., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
17. Durvalumab and guadecitabine in advanced clear cell renal cell carcinoma: results from the phase Ib/II study BTCRC-GU16-043.
- Author
-
Zakharia Y, Singer EA, Acharyya S, Garje R, Joshi M, Peace D, Baladandayuthapani V, Majumdar A, Li X, Lalancette C, Kryczek I, Zou W, and Alva A
- Subjects
- Humans, Azacitidine therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell genetics, Antibodies, Monoclonal
- Abstract
Epigenetic modulation is well established in hematologic malignancies but to a lesser degree in solid tumors. Here we report the results of a phase Ib/II study of guadecitabine and durvalumab in advanced clear cell renal cell carcinoma (ccRCC; NCT03308396). Patients received guadecitabine (starting at 60 mg/m
2 subcutaneously on days 1-5 with de-escalation to 45 mg/m2 in case of dose limiting toxicity) with durvalumab (1500 mg intravenously on day 8). The study enrolled 57 patients, 6 in phase Ib with safety being the primary objective and 51in phase II, comprising 2 cohorts: 36 patients in Cohort 1 were treatment naive to checkpoint inhibitors (CPI) with 0-1 prior therapies and 15 patients in Cohort 2 were treated with up to two prior systemic therapies including one CPI. The combination of guadecitabine 45 mg/m2 with durvalumab 1500 mg was deemed safe. The primary objective of overall response rate (ORR) in cohort 1 was 22%. Sixteen patients (44%) experienced stable disease (SD). Secondary objectives included overall survival (OS), duration of response, progression-free survival (PFS), clinical benefit rate, and safety as well as ORR for Cohort 2. Median PFS for cohort 1 and cohort 2 were 14.26 and 3.91 months respectively. Median OS was not reached. In cohort 2, one patient achieved a partial response and 60% achieved SD. Asymptomatic neutropenia was the most common adverse event. Even though the trial did not meet the primary objective in cohort 1, the tolerability and PFS signal in CPI naive patients are worth further investigation., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
18. Microbiota-dependent activation of CD4 + T cells induces CTLA-4 blockade-associated colitis via Fcγ receptors.
- Author
-
Lo BC, Kryczek I, Yu J, Vatan L, Caruso R, Matsumoto M, Sato Y, Shaw MH, Inohara N, Xie Y, Lei YL, Zou W, and Núñez G
- Subjects
- Animals, Mice, CTLA-4 Antigen antagonists & inhibitors, Mice, Inbred C57BL, CD4-Positive T-Lymphocytes immunology, Colitis etiology, Colitis microbiology, Microbiota immunology, Receptors, IgG immunology, Lymphocyte Activation, Immune Checkpoint Inhibitors adverse effects
- Abstract
Immune checkpoint inhibitors can stimulate antitumor immunity but can also induce toxicities termed immune-related adverse events (irAEs). Colitis is a common and severe irAE that can lead to treatment discontinuation. Mechanistic understanding of gut irAEs has been hampered because robust colitis is not observed in laboratory mice treated with checkpoint inhibitors. We report here that this limitation can be overcome by using mice harboring the microbiota of wild-caught mice, which develop overt colitis following treatment with anti-CTLA-4 antibodies. Intestinal inflammation is driven by unrestrained activation of IFNγ-producing CD4
+ T cells and depletion of peripherally induced regulatory T cells through Fcγ receptor signaling. Accordingly, anti-CTLA-4 nanobodies that lack an Fc domain can promote antitumor responses without triggering colitis. This work suggests a strategy for mitigating gut irAEs while preserving antitumor stimulating effects of CTLA-4 blockade.- Published
- 2024
- Full Text
- View/download PDF
19. Targeting the lipid kinase PIKfyve upregulates surface expression of MHC class I to augment cancer immunotherapy.
- Author
-
Bao Y, Qiao Y, Choi JE, Zhang Y, Mannan R, Cheng C, He T, Zheng Y, Yu J, Gondal M, Cruz G, Grove S, Cao X, Su F, Wang R, Chang Y, Kryczek I, Cieslik M, Green MD, Zou W, and Chinnaiyan AM
- Subjects
- Mice, Animals, Humans, Genes, MHC Class I, Histocompatibility Antigens Class I, Immunotherapy methods, Lipids, CD8-Positive T-Lymphocytes, Neoplasms genetics, Neoplasms therapy
- Abstract
Despite the remarkable clinical success of immunotherapies in a subset of cancer patients, many fail to respond to treatment and exhibit resistance. Here, we found that genetic or pharmacologic inhibition of the lipid kinase PIKfyve, a regulator of autophagic flux and lysosomal biogenesis, upregulated surface expression of major histocompatibility complex class I (MHC-I) in cancer cells via impairing autophagic flux, resulting in enhanced cancer cell killing mediated by CD8
+ T cells. Genetic depletion or pharmacologic inhibition of PIKfyve elevated tumor-specific MHC-I surface expression, increased intratumoral functional CD8+ T cells, and slowed tumor progression in multiple syngeneic mouse models. Importantly, enhanced antitumor responses by Pikfyve -depletion were CD8+ T cell- and MHC-I-dependent, as CD8+ T cell depletion or B2m knockout rescued tumor growth. Furthermore, PIKfyve inhibition improved response to immune checkpoint blockade (ICB), adoptive cell therapy, and a therapeutic vaccine. High expression of PIKFYVE was also predictive of poor response to ICB and prognostic of poor survival in ICB-treated cohorts. Collectively, our findings show that targeting PIKfyve enhances immunotherapies by elevating surface expression of MHC-I in cancer cells, and PIKfyve inhibitors have potential as agents to increase immunotherapy response in cancer patients., Competing Interests: Competing interests statement:A.M.C. is a co-founder of and serves as a Scientific Advisory Board member for LynxDx, Esanik Therapeutics, Medsyn, and Flamingo Therapeutics. Esanik Therapeutics holds the rights to ESK981 and is developing this compound clinically but was not involved in the design or conduct of this study. A.M.C. is a scientific advisor or consultant for EdenRoc, Proteovant, Belharra, Rappta Therapeutics, and Tempus. W.Z. is a scientific advisor or consultant for NGM Bio, CrownBio, Cstone, ProteoVant, Hengenix, NextCure, and Intergalactic. No competing interests were declared from the remaining authors.- Published
- 2023
- Full Text
- View/download PDF
20. Intersection of immune and oncometabolic pathways drives cancer hyperprogression during immunotherapy.
- Author
-
Li G, Choi JE, Kryczek I, Sun Y, Liao P, Li S, Wei S, Grove S, Vatan L, Nelson R, Schaefer G, Allen SG, Sankar K, Fecher LA, Mendiratta-Lala M, Frankel TL, Qin A, Waninger JJ, Tezel A, Alva A, Lao CD, Ramnath N, Cieslik M, Harms PW, Green MD, Chinnaiyan AM, and Zou W
- Subjects
- Animals, CD8-Positive T-Lymphocytes, Fibroblast Growth Factor 2, Disease Progression, Interferon-gamma, Immunotherapy methods, beta Catenin, Neoplasms therapy, Neoplasms pathology
- Abstract
Immune checkpoint blockade (ICB) can produce durable responses against cancer. We and others have found that a subset of patients experiences paradoxical rapid cancer progression during immunotherapy. It is poorly understood how tumors can accelerate their progression during ICB. In some preclinical models, ICB causes hyperprogressive disease (HPD). While immune exclusion drives resistance to ICB, counterintuitively, patients with HPD and complete response (CR) following ICB manifest comparable levels of tumor-infiltrating CD8
+ T cells and interferon γ (IFNγ) gene signature. Interestingly, patients with HPD but not CR exhibit elevated tumoral fibroblast growth factor 2 (FGF2) and β-catenin signaling. In animal models, T cell-derived IFNγ promotes tumor FGF2 signaling, thereby suppressing PKM2 activity and decreasing NAD+ , resulting in reduction of SIRT1-mediated β-catenin deacetylation and enhanced β-catenin acetylation, consequently reprograming tumor stemness. Targeting the IFNγ-PKM2-β-catenin axis prevents HPD in preclinical models. Thus, the crosstalk of core immunogenic, metabolic, and oncogenic pathways via the IFNγ-PKM2-β-catenin cascade underlies ICB-associated HPD., Competing Interests: Declaration of interests W.Z. has served as a scientific advisor or consultant for NGM, CrownBio, Cstone, ProteoVant, Hengenix, NextCure, and Intergalactic. L.A.F receives clinical trial support from Array, Kartos, BMS, EMD Serono, and Pfizer and is a consultant for Elsevier. C.D.L. receives clinical trial support from BMS, Merck, and Novartis. A.Q. has research funding from Merck and Clovis. A.A. serves as a consultant for Merck, AstraZeneca, Bristol-Myers Squibb, and Pfizer/EMD Serono. A.A. receives research funding through the University of Michigan from Merck, Genentech, Prometheus Laboratories, Mirati Therapeutics, Roche, Bayer, Progenics, Astellas Pharma, Arcus Biosciences, AstraZeneca, Bristol-Myers Squibb, and Clovis Oncology., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
21. OXPHOS promotes apoptotic resistance and cellular persistence in T H 17 cells in the periphery and tumor microenvironment.
- Author
-
Hong HS, Mbah NE, Shan M, Loesel K, Lin L, Sajjakulnukit P, Correa LO, Andren A, Lin J, Hayashi A, Magnuson B, Chen J, Li Z, Xie Y, Zhang L, Goldstein DR, Carty SA, Lei YL, Opipari AW, Argüello RJ, Kryczek I, Kamada N, Zou W, Franchi L, and Lyssiotis CA
- Subjects
- Mice, Animals, Mitochondria metabolism, Glycolysis genetics, Cell Differentiation, Oxidative Phosphorylation, Tumor Microenvironment
- Abstract
T cell proliferation and cytokine production are bioenergetically and biosynthetically costly. The inability to meet these metabolic demands results in altered differentiation, accompanied by impaired effector function, and attrition of the immune response. Interleukin-17-producing CD4 T cells (T
H 17s) are mediators of host defense, autoimmunity, and antitumor immunity in the setting of adoptive T cell therapy. TH 17s are long-lived cells that require mitochondrial oxidative phosphorylation (OXPHOS) for effector function in vivo. Considering that TH 17s polarized under standardized culture conditions are predominately glycolytic, little is known about how OXPHOS regulates TH 17 processes, such as their ability to persist and thus contribute to protracted immune responses. Here, we modified standardized culture medium and identified a culture system that reliably induces OXPHOS dependence in TH 17s. We found that TH 17s cultured under OXPHOS conditions metabolically resembled their in vivo counterparts, whereas glycolytic cultures were dissimilar. OXPHOS TH 17s exhibited increased mitochondrial fitness, glutamine anaplerosis, and an antiapoptotic phenotype marked by high BCL-XL and low BIM. Limited mitophagy, mediated by mitochondrial fusion regulator OPA-1, was critical to apoptotic resistance in OXPHOS TH 17s. By contrast, glycolytic TH 17s exhibited more mitophagy and an imbalance in BCL-XL to BIM, thereby priming them for apoptosis. In addition, through adoptive transfer experiments, we demonstrated that OXPHOS protected TH 17s from apoptosis while enhancing their persistence in the periphery and tumor microenvironment in a murine model of melanoma. Together, our work demonstrates how metabolism regulates TH 17 cell fate and highlights the potential for therapies that target OXPHOS in TH 17-driven diseases.- Published
- 2022
- Full Text
- View/download PDF
22. CD8 + T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4.
- Author
-
Liao P, Wang W, Wang W, Kryczek I, Li X, Bian Y, Sell A, Wei S, Grove S, Johnson JK, Kennedy PD, Gijón M, Shah YM, and Zou W
- Subjects
- CD8-Positive T-Lymphocytes metabolism, Coenzyme A Ligases metabolism, Fatty Acids, Humans, Ferroptosis, Neoplasms
- Abstract
Tumor cell intrinsic ferroptosis-initiating mechanisms are unknown. Here, we discover that T cell-derived interferon (IFN)γ in combination with arachidonic acid (AA) induces immunogenic tumor ferroptosis, serving as a mode of action for CD8
+ T cell (CTL)-mediated tumor killing. Mechanistically, IFNγ stimulates ACSL4 and alters tumor cell lipid pattern, thereby increasing incorporations of AA into C16 and C18 acyl chain-containing phospholipids. Palmitoleic acid and oleic acid, two common C16 and C18 fatty acids in blood, promote ACSL4-dependent tumor ferroptosis induced by IFNγ plus AA. Moreover, tumor ACSL4 deficiency accelerates tumor progression. Low-dose AA enhances tumor ferroptosis and elevates spontaneous and immune checkpoint blockade (ICB)-induced anti-tumor immunity. Clinically, tumor ACSL4 correlates with T cell signatures and improved survival in ICB-treated cancer patients. Thus, IFNγ signaling paired with selective fatty acids is a natural tumor ferroptosis-promoting mechanism and a mode of action of CTLs. Targeting the ACSL4 pathway is a potential anti-cancer approach., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
23. Metabolism drives macrophage heterogeneity in the tumor microenvironment.
- Author
-
Li S, Yu J, Huber A, Kryczek I, Wang Z, Jiang L, Li X, Du W, Li G, Wei S, Vatan L, Szeliga W, Chinnaiyan AM, Green MD, Cieslik M, and Zou W
- Subjects
- Animals, Gene Expression Profiling, Macrophages metabolism, Mice, Tumor-Associated Macrophages, Liver Neoplasms pathology, Tumor Microenvironment
- Abstract
Tumor-associated macrophages (TAMs) are a major cellular component in the tumor microenvironment (TME). However, the relationship between the phenotype and metabolic pattern of TAMs remains poorly understood. We performed single-cell transcriptome profiling on hepatic TAMs from mice bearing liver metastatic tumors. We find that TAMs manifest high heterogeneity at the levels of transcription, development, metabolism, and function. Integrative analyses and validation experiments indicate that increased purine metabolism is a feature of TAMs with pro-tumor and terminal differentiation phenotypes. Like mouse TAMs, human TAMs are highly heterogeneous. Human TAMs with increased purine metabolism exhibit a pro-tumor phenotype and correlate with poor therapeutic efficacy to immune checkpoint blockade. Altogether, our work demonstrates that TAMs are developmentally, metabolically, and functionally heterogeneous and purine metabolism may be a key metabolic feature of a pro-tumor macrophage population., Competing Interests: Declaration of interests We declare that we have no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. DOT1L affects colorectal carcinogenesis via altering T cell subsets and oncogenic pathway.
- Author
-
Sun D, Wang W, Guo F, Pitter MR, Du W, Wei S, Grove S, Vatan L, Chen Y, Kryczek I, Fearon ER, Fang JY, and Zou W
- Subjects
- Carcinogenesis metabolism, Forkhead Transcription Factors metabolism, Humans, Inflammation, Tumor Microenvironment, Wnt Signaling Pathway, beta Catenin genetics, beta Catenin metabolism, Colorectal Neoplasms pathology, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, T-Lymphocyte Subsets metabolism, T-Lymphocyte Subsets pathology
- Abstract
Chronic inflammation and oncogenic pathway activation are key-contributing factors in colorectal cancer pathogenesis. However, colorectal intrinsic mechanisms linking these two factors in cancer development are poorly defined. Here, we show that intestinal epithelial cell (IEC)-specific deletion of Dot1l histone methyltransferase ( Dot1l
ΔIEC ) reduced H3K79 dimethylation (H3K79me2) in IECs and inhibited intestinal tumor formation in ApcMin - and AOM-DSS-induced colorectal cancer models. IEC- Dot1l abrogation was accompanied by alleviative colorectal inflammation and reduced Wnt/β-catenin signaling activation. Mechanistically, Dot1l deficiency resulted in an increase in Foxp3+ RORϒ+ regulatory T (Treg) cells and a decrease in inflammatory Th17 and Th22 cells, thereby reducing local inflammation in the intestinal tumor microenvironment. Furthermore, Dot1l deficiency caused a reduction of H3K79me2 occupancies in the promoters of the Wnt/β-catenin signaling genes, thereby diminishing Wnt/β-catenin oncogenic signaling pathway activation in colorectal cancer cells. Clinically, high levels of tumor H3K79me2 were detected in patients with colorectal carcinomas as compared to adenomas, and negatively correlated with RORϒ+ FOXP3+ Treg cells. Altogether, we conclude that DOT1L is an intrinsic molecular node connecting chronic immune activation and oncogenic signaling pathways in colorectal cancer. Our work suggests that targeting the DOT1L pathway may control colorectal carcinogenesis. Significance : IEC-intrinsic DOT1L controls T cell subset balance and key oncogenic pathway activation, impacting colorectal carcinogenesis., Competing Interests: No potential conflict of interest was reported by the author(s)., (© 2022 The Author(s). Published with license by Taylor & Francis Group, LLC.)- Published
- 2022
- Full Text
- View/download PDF
25. Autophagy Inhibition by Targeting PIKfyve Potentiates Response to Immune Checkpoint Blockade in Prostate Cancer.
- Author
-
Qiao Y, Choi JE, Tien JC, Simko SA, Rajendiran T, Vo JN, Delekta AD, Wang L, Xiao L, Hodge NB, Desai P, Mendoza S, Juckette K, Xu A, Soni T, Su F, Wang R, Cao X, Yu J, Kryczek I, Wang XM, Wang X, Siddiqui J, Wang Z, Bernard A, Fernandez-Salas E, Navone NM, Ellison SJ, Ding K, Eskelinen EL, Heath EI, Klionsky DJ, Zou W, and Chinnaiyan AM
- Subjects
- Autophagy, Humans, Immunotherapy methods, Male, Phosphatidylinositol 3-Kinases pharmacology, Tumor Microenvironment, Immune Checkpoint Inhibitors, Prostatic Neoplasms, Castration-Resistant drug therapy
- Abstract
Multi-tyrosine kinase inhibitors (MTKIs) have thus far had limited success in the treatment of castration-resistant prostate cancer (CRPC). Here, we report a phase I-cleared orally bioavailable MTKI, ESK981, with a novel autophagy inhibitory property that decreased tumor growth in diverse preclinical models of CRPC. The anti-tumor activity of ESK981 was maximized in immunocompetent tumor environments where it upregulated CXCL10 expression through the interferon gamma pathway and promoted functional T cell infiltration, which resulted in enhanced therapeutic response to immune checkpoint blockade. Mechanistically, we identify the lipid kinase PIKfyve as the direct target of ESK981. PIKfyve-knockdown recapitulated ESK981's anti-tumor activity and enhanced the therapeutic benefit of immune checkpoint blockade. Our study reveals that targeting PIKfyve via ESK981 turns tumors from cold into hot through inhibition of autophagy, which may prime the tumor immune microenvironment in advanced prostate cancer patients and be an effective treatment strategy alone or in combination with immunotherapies.
- Published
- 2021
- Full Text
- View/download PDF
26. IFNα Augments Clinical Efficacy of Regulatory T-cell Depletion with Denileukin Diftitox in Ovarian Cancer.
- Author
-
Thibodeaux SR, Barnett BB, Pandeswara S, Wall SR, Hurez V, Dao V, Sun L, Daniel BJ, Brumlik MJ, Drerup J, Padrón Á, Whiteside T, Kryczek I, Zou W, and Curiel TJ
- Subjects
- Animals, Drug Therapy, Combination, Female, Humans, Mice, Recombinant Fusion Proteins therapeutic use, Treatment Outcome, Tumor Cells, Cultured, Antineoplastic Agents therapeutic use, Diphtheria Toxin therapeutic use, Interferon-alpha therapeutic use, Interleukin-2 therapeutic use, Lymphocyte Depletion, Ovarian Neoplasms drug therapy, T-Lymphocytes, Regulatory
- Abstract
Purpose: Immunotherapy treats some cancers, but not ovarian cancer. Regulatory T cells (Tregs) impede anti-ovarian cancer immunity but effective human Treg-directed treatments are lacking. We tested Treg depletion with denileukin diftitox (DD) ± IFNα as ovarian cancer immunotherapy., Patients and Methods: Mice with syngeneic ID8 ovarian cancer challenge were treated with DD, IFNα, or both. The phase 0/I trial tested one dose-escalated DD infusion for functional Treg reduction, safety, and tolerability. The phase II trial added IFNα2a to DD if DD alone failed clinically., Results: DD depleted Tregs, and improved antitumor immunity and survival in mice. IFNα significantly improved antitumor immunity and survival with DD. IFNα did not alter Treg numbers or function but boosted tumor-specific immunity and reduced tumor Treg function with DD by inducing dendritic cell IL6. DD alone was well tolerated, depleted functional blood Tregs and improved immunity in patients with various malignancies in phase 0/I. A patient with ovarian cancer in phase 0/I experienced partial clinical response prompting a phase II ovarian cancer trial, but DD alone failed phase II. Another phase II trial added pegylated IFNα2a to failed DD, producing immunologic and clinical benefit in two of two patients before a DD shortage halt. DD alone was well tolerated. Adding IFNα increased toxicities but was tolerable, and reduced human Treg numbers in blood, and function through dendritic cell-induced IL6 in vitro ., Conclusions: Treg depletion is clinically useful but unlikely alone to cure ovarian cancer. Rational treatment agent combinations can salvage clinical failure of Treg depletion alone, even when neither single agent provides meaningful clinical benefit., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
27. Loss of Optineurin Drives Cancer Immune Evasion via Palmitoylation-Dependent IFNGR1 Lysosomal Sorting and Degradation.
- Author
-
Du W, Hua F, Li X, Zhang J, Li S, Wang W, Zhou J, Wang W, Liao P, Yan Y, Li G, Wei S, Grove S, Vatan L, Zgodziński W, Majewski M, Wallner G, Chen H, Kryczek I, Fang JY, and Zou W
- Subjects
- Animals, Colorectal Neoplasms genetics, Colorectal Neoplasms therapy, Female, Histocompatibility Antigens Class I metabolism, Humans, Interferon-gamma metabolism, Lipoylation, Male, Mice, Mice, Inbred C57BL, Mice, Inbred Strains, Protein Transport, Specific Pathogen-Free Organisms, Interferon gamma Receptor, Cell Cycle Proteins metabolism, Colorectal Neoplasms metabolism, Membrane Transport Proteins metabolism, Receptors, Interferon metabolism
- Abstract
Mutations in IFN and MHC signaling genes endow immunotherapy resistance. Patients with colorectal cancer infrequently exhibit IFN and MHC signaling gene mutations and are generally resistant to immunotherapy. In exploring the integrity of IFN and MHC signaling in colorectal cancer, we found that optineurin was a shared node between the two pathways and predicted colorectal cancer patient outcome. Loss of optineurin occurs in early-stage human colorectal cancer. Immunologically, optineurin deficiency was shown to attenuate IFNGR1 and MHC-I expression, impair T-cell immunity, and diminish immunotherapy efficacy in murine cancer models and patients with cancer. Mechanistically, we observed that IFNGR1 was S -palmitoylated on Cys122, and AP3D1 bound with and sorted palmitoylated IFNGR1 to lysosome for degradation. Unexpectedly, optineurin interacted with AP3D1 to prevent palmitoylated IFNGR1 lysosomal sorting and degradation, thereby maintaining IFNγ and MHC-I signaling integrity. Furthermore, pharmacologically targeting IFNGR1 palmitoylation stabilized IFNGR1, augmented tumor immunity, and sensitized checkpoint therapy. Thus, loss of optineurin drives immune evasion and intrinsic immunotherapy resistance in colorectal cancer. SIGNIFICANCE: Loss of optineurin impairs the integrity of both IFNγ and MHC-I signaling pathways via palmitoylation-dependent IFNGR1 lysosomal sorting and degradation, thereby driving immune evasion and intrinsic immunotherapy resistance in colorectal cancer. Our work suggests that pharmacologically targeting IFNGR1 palmitoylation can stabilize IFNGR1, enhance T-cell immunity, and sensitize checkpoint therapy in colorectal cancer. See related commentary by Salvagno and Cubillos-Ruiz, p. 1623 . This article is highlighted in the In This Issue feature, p. 1601 ., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
28. LIMIT is an immunogenic lncRNA in cancer immunity and immunotherapy.
- Author
-
Li G, Kryczek I, Nam J, Li X, Li S, Li J, Wei S, Grove S, Vatan L, Zhou J, Du W, Lin H, Wang T, Subramanian C, Moon JJ, Cieslik M, Cohen M, and Zou W
- Subjects
- Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, HSP90 Heat-Shock Proteins metabolism, Humans, Neoplasms immunology, Signal Transduction physiology, Immunotherapy methods, Neoplasms drug therapy, Neoplasms metabolism, RNA, Long Noncoding genetics
- Abstract
Major histocompatibility complex-I (MHC-I) presents tumour antigens to CD8
+ T cells and triggers anti-tumour immunity. Humans may have 30,000-60,000 long noncoding RNAs (lncRNAs). However, it remains poorly understood whether lncRNAs affect tumour immunity. Here, we identify a lncRNA, lncRNA inducing MHC-I and immunogenicity of tumour (LIMIT), in humans and mice. We found that IFNγ stimulated LIMIT, LIMIT cis-activated the guanylate-binding protein (GBP) gene cluster and GBPs disrupted the association between HSP90 and heat shock factor-1 (HSF1), thereby resulting in HSF1 activation and transcription of MHC-I machinery, but not PD-L1. RNA-guided CRISPR activation of LIMIT boosted GBPs and MHC-I, and potentiated tumour immunogenicity and checkpoint therapy. Silencing LIMIT, GBPs and/or HSF1 diminished MHC-I, impaired antitumour immunity and blunted immunotherapy efficacy. Clinically, LIMIT, GBP- and HSF1-signalling transcripts and proteins correlated with MHC-I, tumour-infiltrating T cells and checkpoint blockade response in patients with cancer. Together, we demonstrate that LIMIT is a cancer immunogenic lncRNA and the LIMIT-GBP-HSF1 axis may be targetable for cancer immunotherapy.- Published
- 2021
- Full Text
- View/download PDF
29. Stanniocalcin 1 is a phagocytosis checkpoint driving tumor immune resistance.
- Author
-
Lin H, Kryczek I, Li S, Green MD, Ali A, Hamasha R, Wei S, Vatan L, Szeliga W, Grove S, Li X, Li J, Wang W, Yan Y, Choi JE, Li G, Bian Y, Xu Y, Zhou J, Yu J, Xia H, Wang W, Alva A, Chinnaiyan AM, Cieslik M, and Zou W
- Subjects
- Animals, Antigen Presentation immunology, Immunotherapy methods, Macrophages metabolism, Mice, Phagocytosis drug effects, Receptors, Immunologic immunology, Glycoproteins metabolism, Lymphocyte Activation immunology, Macrophages immunology, Phagocytosis immunology, Tumor Escape immunology
- Abstract
Immunotherapy induces durable clinical responses in a fraction of patients with cancer. However, therapeutic resistance poses a major challenge to current immunotherapies. Here, we identify that expression of tumor stanniocalcin 1 (STC1) correlates with immunotherapy efficacy and is negatively associated with patient survival across diverse cancer types. Gain- and loss-of-function experiments demonstrate that tumor STC1 supports tumor progression and enables tumor resistance to checkpoint blockade in murine tumor models. Mechanistically, tumor STC1 interacts with calreticulin (CRT), an "eat-me" signal, and minimizes CRT membrane exposure, thereby abrogating membrane CRT-directed phagocytosis by antigen-presenting cells (APCs), including macrophages and dendritic cells. Consequently, this impairs APC capacity of antigen presentation and T cell activation. Thus, tumor STC1 inhibits APC phagocytosis and contributes to tumor immune evasion and immunotherapy resistance. We suggest that STC1 is a previously unappreciated phagocytosis checkpoint and targeting STC1 and its interaction with CRT may sensitize to cancer immunotherapy., Competing Interests: Declaration of interests The authors declare no conflict of interest., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. The ubiquitin ligase MDM2 sustains STAT5 stability to control T cell-mediated antitumor immunity.
- Author
-
Zhou J, Kryczek I, Li S, Li X, Aguilar A, Wei S, Grove S, Vatan L, Yu J, Yan Y, Liao P, Lin H, Li J, Li G, Du W, Wang W, Lang X, Wang W, Wang S, and Zou W
- Subjects
- Animals, Antineoplastic Agents pharmacology, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes transplantation, Cell Line, Tumor, Combined Modality Therapy, Female, Gene Expression Regulation, Neoplastic, HEK293 Cells, Humans, Immunotherapy, Adoptive, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating transplantation, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Neoplasms genetics, Neoplasms immunology, Neoplasms therapy, Protein Stability, Proteolysis, Proto-Oncogene Proteins c-mdm2 genetics, STAT5 Transcription Factor genetics, Signal Transduction, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, CD8-Positive T-Lymphocytes enzymology, Lymphocytes, Tumor-Infiltrating enzymology, Neoplasms enzymology, Proto-Oncogene Proteins c-mdm2 metabolism, STAT5 Transcription Factor metabolism
- Abstract
Targeting the p53-MDM2 pathway to reactivate tumor p53 is a chemotherapeutic approach. However, the involvement of this pathway in CD8
+ T cell-mediated antitumor immunity is unknown. Here, we report that mice with MDM2 deficiency in T cells exhibit accelerated tumor progression and a decrease in tumor-infiltrating CD8+ T cell survival and function. Mechanistically, MDM2 competes with c-Cbl for STAT5 binding, reduces c-Cbl-mediated STAT5 degradation and enhances STAT5 stability in tumor-infiltrating CD8+ T cells. Targeting the p53-MDM2 interaction with a pharmacological agent, APG-115, augmented MDM2 in T cells, thereby stabilizing STAT5, boosting T cell immunity and synergizing with cancer immunotherapy. Unexpectedly, these effects of APG-115 were dependent on p53 and MDM2 in T cells. Clinically, MDM2 abundance correlated with T cell function and interferon-γ signature in patients with cancer. Thus, the p53-MDM2 pathway controls T cell immunity, and targeting this pathway may treat patients with cancer regardless of tumor p53 status.- Published
- 2021
- Full Text
- View/download PDF
31. Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T cell elimination.
- Author
-
Yu J, Green MD, Li S, Sun Y, Journey SN, Choi JE, Rizvi SM, Qin A, Waninger JJ, Lang X, Chopra Z, El Naqa I, Zhou J, Bian Y, Jiang L, Tezel A, Skvarce J, Achar RK, Sitto M, Rosen BS, Su F, Narayanan SP, Cao X, Wei S, Szeliga W, Vatan L, Mayo C, Morgan MA, Schonewolf CA, Cuneo K, Kryczek I, Ma VT, Lao CD, Lawrence TS, Ramnath N, Wen F, Chinnaiyan AM, Cieslik M, Alva A, and Zou W
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung immunology, Carcinoma, Non-Small-Cell Lung secondary, Carcinoma, Non-Small-Cell Lung therapy, Cell Line, Tumor, Cohort Studies, Combined Modality Therapy, Female, Humans, Liver Neoplasms immunology, Liver Neoplasms, Experimental immunology, Lymphocyte Activation, Male, Melanoma immunology, Melanoma secondary, Melanoma therapy, Mice, Mice, Inbred C57BL, Mice, Transgenic, Radiotherapy, Adjuvant, T-Lymphocytes classification, T-Lymphocytes pathology, Treatment Failure, Treatment Outcome, Tumor Microenvironment immunology, Tumor Microenvironment radiation effects, Immunotherapy, Liver Neoplasms secondary, Liver Neoplasms therapy, Liver Neoplasms, Experimental secondary, Liver Neoplasms, Experimental therapy, Macrophages immunology, T-Lymphocytes immunology
- Abstract
Metastasis is the primary cause of cancer mortality, and cancer frequently metastasizes to the liver. It is not clear whether liver immune tolerance mechanisms contribute to cancer outcomes. We report that liver metastases diminish immunotherapy efficacy systemically in patients and preclinical models. Patients with liver metastases derive limited benefit from immunotherapy independent of other established biomarkers of response. In multiple mouse models, we show that liver metastases siphon activated CD8
+ T cells from systemic circulation. Within the liver, activated antigen-specific Fas+ CD8+ T cells undergo apoptosis following their interaction with FasL+ CD11b+ F4/80+ monocyte-derived macrophages. Consequently, liver metastases create a systemic immune desert in preclinical models. Similarly, patients with liver metastases have reduced peripheral T cell numbers and diminished tumoral T cell diversity and function. In preclinical models, liver-directed radiotherapy eliminates immunosuppressive hepatic macrophages, increases hepatic T cell survival and reduces hepatic siphoning of T cells. Thus, liver metastases co-opt host peripheral tolerance mechanisms to cause acquired immunotherapy resistance through CD8+ T cell deletion, and the combination of liver-directed radiotherapy and immunotherapy could promote systemic antitumor immunity.- Published
- 2021
- Full Text
- View/download PDF
32. Autophagic adaptation to oxidative stress alters peritoneal residential macrophage survival and ovarian cancer metastasis.
- Author
-
Xia H, Li S, Li X, Wang W, Bian Y, Wei S, Grove S, Wang W, Vatan L, Liu JR, McLean K, Rattan R, Munkarah A, Guan JL, Kryczek I, and Zou W
- Subjects
- Adaptation, Physiological, Animals, Autophagy-Related Proteins physiology, Female, Humans, Leukocyte Common Antigens physiology, Macrophages, Peritoneal metabolism, Membrane Proteins genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Ovarian Neoplasms metabolism, Peritoneal Neoplasms metabolism, Receptors, CCR2 physiology, Autophagy, Macrophages, Peritoneal pathology, Membrane Proteins metabolism, Membrane Proteins physiology, Ovarian Neoplasms pathology, Oxidative Stress, Peritoneal Neoplasms secondary
- Abstract
Tumor-associated macrophages (TAMs) affect cancer progression and therapy. Ovarian carcinoma often metastasizes to the peritoneal cavity. Here, we found 2 peritoneal macrophage subsets in mice bearing ID8 ovarian cancer based on T cell immunoglobulin and mucin domain containing 4 (Tim-4) expression. Tim-4+ TAMs were embryonically originated and locally sustained while Tim-4- TAMs were replenished from circulating monocytes. Tim-4+ TAMs, but not Tim-4- TAMs, promoted tumor growth in vivo. Relative to Tim-4- TAMs, Tim-4+ TAMs manifested high oxidative phosphorylation and adapted mitophagy to alleviate oxidative stress. High levels of arginase-1 in Tim-4+ TAMs contributed to potent mitophagy activities via weakened mTORC1 activation due to low arginine resultant from arginase-1-mediated metabolism. Furthermore, genetic deficiency of autophagy element FAK family-interacting protein of 200 kDa resulted in Tim-4+ TAM loss via ROS-mediated apoptosis and elevated T cell immunity and ID8 tumor inhibition in vivo. Moreover, human ovarian cancer-associated macrophages positive for complement receptor of the immunoglobulin superfamily (CRIg) were transcriptionally, metabolically, and functionally similar to murine Tim-4+ TAMs. Thus, targeting CRIg+ (Tim-4+) TAMs may potentially treat patients with ovarian cancer with peritoneal metastasis.
- Published
- 2020
- Full Text
- View/download PDF
33. Cancer SLC43A2 alters T cell methionine metabolism and histone methylation.
- Author
-
Bian Y, Li W, Kremer DM, Sajjakulnukit P, Li S, Crespo J, Nwosu ZC, Zhang L, Czerwonka A, Pawłowska A, Xia H, Li J, Liao P, Yu J, Vatan L, Szeliga W, Wei S, Grove S, Liu JR, McLean K, Cieslik M, Chinnaiyan AM, Zgodziński W, Wallner G, Wertel I, Okła K, Kryczek I, Lyssiotis CA, and Zou W
- Subjects
- Amino Acid Transport System L deficiency, Animals, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Epigenesis, Genetic, Female, Histones chemistry, Humans, Mice, Neoplasms genetics, Neoplasms immunology, Neoplasms pathology, Receptors, Antigen, T-Cell metabolism, STAT5 Transcription Factor metabolism, Amino Acid Transport System L metabolism, CD8-Positive T-Lymphocytes metabolism, Histones metabolism, Methionine metabolism, Methylation, Neoplasms metabolism
- Abstract
Abnormal epigenetic patterns correlate with effector T cell malfunction in tumours
1-4 , but the cause of this link is unknown. Here we show that tumour cells disrupt methionine metabolism in CD8+ T cells, thereby lowering intracellular levels of methionine and the methyl donor S-adenosylmethionine (SAM) and resulting in loss of dimethylation at lysine 79 of histone H3 (H3K79me2). Loss of H3K79me2 led to low expression of STAT5 and impaired T cell immunity. Mechanistically, tumour cells avidly consumed methionine and outcompeted T cells for methionine by expressing high levels of the methionine transporter SLC43A2. Genetic and biochemical inhibition of tumour SLC43A2 restored H3K79me2 in T cells, thereby boosting spontaneous and checkpoint-induced tumour immunity. Moreover, methionine supplementation improved the expression of H3K79me2 and STAT5 in T cells, and this was accompanied by increased T cell immunity in tumour-bearing mice and patients with colon cancer. Clinically, tumour SLC43A2 correlated negatively with T cell histone methylation and functional gene signatures. Our results identify a mechanistic connection between methionine metabolism, histone patterns, and T cell immunity in the tumour microenvironment. Thus, cancer methionine consumption is an immune evasion mechanism, and targeting cancer methionine signalling may provide an immunotherapeutic approach.- Published
- 2020
- Full Text
- View/download PDF
34. Epigenetic driver mutations in ARID1A shape cancer immune phenotype and immunotherapy.
- Author
-
Li J, Wang W, Zhang Y, Cieślik M, Guo J, Tan M, Green MD, Wang W, Lin H, Li W, Wei S, Zhou J, Li G, Jing X, Vatan L, Zhao L, Bitler B, Zhang R, Cho KR, Dou Y, Kryczek I, Chan TA, Huntsman D, Chinnaiyan AM, and Zou W
- Subjects
- Animals, Cell Line, Tumor, Chromatin Assembly and Disassembly genetics, Chromatin Assembly and Disassembly immunology, DNA-Binding Proteins chemistry, Enhancer of Zeste Homolog 2 Protein chemistry, Enhancer of Zeste Homolog 2 Protein immunology, Epigenesis, Genetic, Female, Humans, Immunophenotyping, Immunotherapy, Interferons genetics, Interferons immunology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating pathology, Melanoma genetics, Melanoma immunology, Melanoma pathology, Mice, Neoplasms pathology, Ovarian Neoplasms genetics, Ovarian Neoplasms immunology, Ovarian Neoplasms pathology, Signal Transduction genetics, Signal Transduction immunology, Transcription Factors chemistry, Tumor Escape genetics, Tumor Escape immunology, DNA-Binding Proteins genetics, DNA-Binding Proteins immunology, Mutation, Neoplasms genetics, Neoplasms immunology, Transcription Factors genetics, Transcription Factors immunology
- Abstract
Whether mutations in cancer driver genes directly affect cancer immune phenotype and T cell immunity remains a standing question. ARID1A is a core member of the polymorphic BRG/BRM-associated factor chromatin remodeling complex. ARID1A mutations occur in human cancers and drive cancer development. Here, we studied the molecular, cellular, and clinical impact of ARID1A aberrations on cancer immunity. We demonstrated that ARID1A aberrations resulted in limited chromatin accessibility to IFN-responsive genes, impaired IFN gene expression, anemic T cell tumor infiltration, poor tumor immunity, and shortened host survival in many human cancer histologies and in murine cancer models. Impaired IFN signaling was associated with poor immunotherapy response. Mechanistically, ARID1A interacted with EZH2 via its carboxyl terminal and antagonized EZH2-mediated IFN responsiveness. Thus, the interaction between ARID1A and EZH2 defines cancer IFN responsiveness and immune evasion. Our work indicates that cancer epigenetic driver mutations can shape cancer immune phenotype and immunotherapy.
- Published
- 2020
- Full Text
- View/download PDF
35. Interleukin 22 Signaling Regulates Acinar Cell Plasticity to Promote Pancreatic Tumor Development in Mice.
- Author
-
Perusina Lanfranca M, Zhang Y, Girgis A, Kasselman S, Lazarus J, Kryczek I, Delrosario L, Rhim A, Koneva L, Sartor M, Sun L, Halbrook C, Nathan H, Shi J, Crawford HC, Pasca di Magliano M, Zou W, and Frankel TL
- Subjects
- Acinar Cells immunology, Animals, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal mortality, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor transplantation, Cell Plasticity drug effects, Cell Plasticity immunology, Cell Transformation, Neoplastic drug effects, Disease Models, Animal, Epithelial-Mesenchymal Transition drug effects, Epithelial-Mesenchymal Transition immunology, Female, HEK293 Cells, Humans, Interleukins immunology, Janus Kinases antagonists & inhibitors, Janus Kinases metabolism, Male, Metaplasia immunology, Metaplasia pathology, Mice, Mice, Knockout, Nitriles, Pancreas cytology, Pancreas immunology, Pancreas pathology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Pancreatitis immunology, Pancreatitis pathology, Pyrazoles pharmacology, Pyridones pharmacology, Pyrimidines, Pyrimidinones pharmacology, RNA, Small Interfering metabolism, Receptors, Interleukin metabolism, STAT3 Transcription Factor antagonists & inhibitors, Signal Transduction drug effects, Survival Analysis, Interleukin-22, Acinar Cells pathology, Carcinoma, Pancreatic Ductal immunology, Cell Transformation, Neoplastic immunology, Interleukins metabolism, Pancreatic Neoplasms immunology, STAT3 Transcription Factor metabolism, Signal Transduction immunology
- Abstract
Background & Aims: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy that invades surrounding structures and metastasizes rapidly. Although inflammation is associated with tumor formation and progression, little is known about the mechanisms of this connection. We investigate the effects of interleukin (IL) 22 in the development of pancreatic tumors in mice., Methods: We performed studies with Pdx1-Cre;LSL-Kras
G12D ;Trp53+/- ;Rosa26EYFP/+ (PKCY) mice, which develop pancreatic tumors, and PKCY mice with disruption of IL22 (PKCY Il22-/- mice). Pancreata were collected at different stages of tumor development and analyzed by immunohistochemistry, immunoblotting, real-time polymerase chain reaction, and flow cytometry. Some mice were given cerulean to induce pancreatitis. Pancreatic cancer cell lines (PD2560) were orthotopically injected into C57BL/6 mice or Il22-/- mice, and tumor development was monitored. Pancreatic cells were injected into the tail veins of mice, and lung metastases were quantified. Acini were collected from C57BL/6 mice and resected human pancreata and were cultured. Cell lines and acini cultures were incubated with IL22 and pharmacologic inhibitors, and protein levels were knocked down with small hairpin RNAs. We performed immunohistochemical analyses of 26 PDACs and 5 nonneoplastic pancreas specimens., Results: We observed increased expression of IL22 and the IL22 receptor (IL22R) in the pancreas compared with other tissues in mice; IL22 increased with pancreatitis and tumorigenesis. Flow cytometry indicated that the IL22 was produced primarily by T-helper 22 cells. PKCY Il22-/- mice did not develop precancerous lesions or pancreatic tumors. The addition of IL22 to cultured acinar cells increased their expression of markers of ductal metaplasia; these effects of IL22 were prevented with inhibitors of Janus kinase signaling to signal transducer and activator of transcription (STAT) (ruxolitinib) or mitogen-activated protein kinase kinase (MEK) (trametinib) and with STAT3 knockdown. Pancreatic cells injected into Il22-/- mice formed smaller tumors than those injected into C57BL/6. Incubation of IL22R-expressing PDAC cells with IL22 promoted spheroid formation and invasive activity, resulting in increased expression of stem-associated transcription factors (GATA4, SOX2, SOX17, and NANOG), and increased markers of the epithelial-mesenchymal transition (CDH1, SNAI2, TWIST1, and beta catenin); ruxolitinib blocked these effects. Human PDAC tissues had higher levels of IL22, phosphorylated STAT3, and markers of the epithelial-mesenchymal transition than nonneoplastic tissues. An increased level of STAT3 in IL22R-positive cells was associated with shorter survival times of patients., Conclusions: We found levels of IL22 to be increased during pancreatitis and pancreatic tumor development and to be required for tumor development and progression in mice. IL22 promotes acinar to ductal metaplasia, stem cell features, and increased expression of markers of the epithelial-mesenchymal transition; inhibitors of STAT3 block these effects. Increased expression of IL22 by PDACs is associated with reduced survival times., (Copyright © 2020 AGA Institute. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
36. Epigenetic stabilization of DC and DC precursor classical activation by TNFα contributes to protective T cell polarization.
- Author
-
Eastman AJ, Xu J, Bermik J, Potchen N, den Dekker A, Neal LM, Zhao G, Malachowski A, Schaller M, Kunkel S, Osterholzer JJ, Kryczek I, and Olszewski MA
- Subjects
- Animals, Cellular Reprogramming drug effects, Cryptococcus drug effects, Cryptococcus physiology, Dendritic Cells drug effects, Female, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Immunomodulation drug effects, Lysine metabolism, Methylation, Mice, Inbred CBA, Myeloid-Lymphoid Leukemia Protein metabolism, Phenotype, Promoter Regions, Genetic genetics, Suppression, Genetic drug effects, T-Lymphocytes drug effects, Th1 Cells drug effects, Th1 Cells immunology, Tumor Necrosis Factor-alpha pharmacology, Cell Polarity drug effects, Cytoprotection drug effects, Dendritic Cells metabolism, Epigenesis, Genetic drug effects, T-Lymphocytes cytology, Tumor Necrosis Factor-alpha metabolism
- Abstract
Epigenetic modifications play critical roles in inducing long-lasting immunological memory in innate immune cells, termed trained immunity. Whether similar epigenetic mechanisms regulate dendtritic cell (DC) function to orchestrate development of adaptive immunity remains unknown. We report that DCs matured with IFNγ and TNFα or matured in the lungs during invasive fungal infection with endogenous TNFα acquired a stable TNFα-dependent DC1 program, rendering them resistant to both antigen- and cytokine-induced alternative activation. TNFα-programmed DC1 had increased association of H3K4me3 with DC1 gene promoter regions. Furthermore, MLL1 inhibition blocked TNFα-mediated DC1 phenotype stabilization. During IFI, TNFα-programmed DC1s were required for the development of sustained T
H 1/TH 17 protective immunity, and bone marrow pre-DCs exhibited TNFα-dependent preprogramming, supporting continuous generation of programmed DC1 throughout the infection. TNFα signaling, associated with epigenetic activation of DC1 genes particularly via H3K4me3, critically contributes to generation and sustenance of type 1/17 adaptive immunity and the immune protection against persistent infection., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)- Published
- 2019
- Full Text
- View/download PDF
37. Radiotherapy and Immunotherapy Promote Tumoral Lipid Oxidation and Ferroptosis via Synergistic Repression of SLC7A11.
- Author
-
Lang X, Green MD, Wang W, Yu J, Choi JE, Jiang L, Liao P, Zhou J, Zhang Q, Dow A, Saripalli AL, Kryczek I, Wei S, Szeliga W, Vatan L, Stone EM, Georgiou G, Cieslik M, Wahl DR, Morgan MA, Chinnaiyan AM, Lawrence TS, and Zou W
- Subjects
- Animals, Antineoplastic Agents, Immunological pharmacology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Cell Survival drug effects, Cell Survival radiation effects, Down-Regulation, Ferroptosis drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, Immunotherapy methods, Interferon-gamma, Lipid Metabolism drug effects, Lipid Metabolism radiation effects, Melanoma, Experimental genetics, Mice, Oxidation-Reduction, Sulfasalazine pharmacology, Xenograft Model Antitumor Assays, Amino Acid Transport System y+ genetics, Antineoplastic Agents, Immunological administration & dosage, Melanoma, Experimental therapy, Sulfasalazine administration & dosage
- Abstract
A challenge in oncology is to rationally and effectively integrate immunotherapy with traditional modalities, including radiotherapy. Here, we demonstrate that radiotherapy induces tumor-cell ferroptosis. Ferroptosis agonists augment and ferroptosis antagonists limit radiotherapy efficacy in tumor models. Immunotherapy sensitizes tumors to radiotherapy by promoting tumor-cell ferroptosis. Mechanistically, IFNγ derived from immunotherapy-activated CD8
+ T cells and radiotherapy-activated ATM independently, yet synergistically, suppresses SLC7A11, a unit of the glutamate-cystine antiporter xc- , resulting in reduced cystine uptake, enhanced tumor lipid oxidation and ferroptosis, and improved tumor control. Thus, ferroptosis is an unappreciated mechanism and focus for the development of effective combinatorial cancer therapy. SIGNIFICANCE: This article describes ferroptosis as a previously unappreciated mechanism of action for radiotherapy. Further, it shows that ferroptosis is a novel point of synergy between immunotherapy and radiotherapy. Finally, it nominates SLC7A11, a critical regulator of ferroptosis, as a mechanistic determinant of synergy between radiotherapy and immunotherapy. This article is highlighted in the In This Issue feature, p. 1631 ., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
38. Inhibition of ATM Increases Interferon Signaling and Sensitizes Pancreatic Cancer to Immune Checkpoint Blockade Therapy.
- Author
-
Zhang Q, Green MD, Lang X, Lazarus J, Parsels JD, Wei S, Parsels LA, Shi J, Ramnath N, Wahl DR, Pasca di Magliano M, Frankel TL, Kryczek I, Lei YL, Lawrence TS, Zou W, and Morgan MA
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, Humans, Immunotherapy, Interferons, Mice, Signal Transduction, CD8-Positive T-Lymphocytes, Pancreatic Neoplasms
- Abstract
Combinatorial strategies are needed to overcome the resistance of pancreatic cancer to immune checkpoint blockade (ICB). DNA damage activates the innate immune response and improves ICB efficacy. Because ATM is an apical kinase in the radiation-induced DNA damage response, we investigated the effects of ATM inhibition and radiation on pancreatic tumor immunogenicity. ATM was inhibited through pharmacologic and genetic strategies in human and murine pancreatic cancer models both in vitro and in vivo . Tumor immunogenicity was evaluated after ATM inhibition alone and in combination with radiation by assessing TBK1 and Type I interferon (T1IFN) signaling as well as tumor growth following PD-L1/PD-1 checkpoint inhibition. Inhibition of ATM increased tumoral T1IFN expression in a cGAS/STING-independent, but TBK1- and SRC-dependent, manner. The combination of ATM inhibition with radiation further enhanced TBK1 activity, T1IFN production, and antigen presentation. Furthermore, ATM silencing increased PD-L1 expression and increased the sensitivity of pancreatic tumors to PD-L1-blocking antibody in association with increased tumoral CD8
+ T cells and established immune memory. In patient pancreatic tumors, low ATM expression inversely correlated with PD-L1 expression. Taken together, these results demonstrate that the efficacy of ICB in pancreatic cancer is enhanced by ATM inhibition and further potentiated by radiation as a function of increased tumoral immunogenicity, underscoring the potential of ATM inhibition in combination with ICB and radiation as an efficacious treatment strategy for pancreatic cancer. SIGNIFICANCE: This study demonstrates that ATM inhibition induces a T1IFN-mediated innate immune response in pancreatic cancer that is further enhanced by radiation and leads to increased sensitivity to anti-PD-L1 therapy. See related commentary by Gutiontov and Weichselbaum, p. 3815 ., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
39. CD8 + T cells regulate tumour ferroptosis during cancer immunotherapy.
- Author
-
Wang W, Green M, Choi JE, Gijón M, Kennedy PD, Johnson JK, Liao P, Lang X, Kryczek I, Sell A, Xia H, Zhou J, Li G, Li J, Li W, Wei S, Vatan L, Zhang H, Szeliga W, Gu W, Liu R, Lawrence TS, Lamb C, Tanno Y, Cieslik M, Stone E, Georgiou G, Chan TA, Chinnaiyan A, and Zou W
- Subjects
- Amino Acid Transport System y+ metabolism, Animals, B7-H1 Antigen antagonists & inhibitors, Cell Line, Tumor, Cysteine metabolism, Female, Fusion Regulatory Protein 1, Heavy Chain metabolism, Humans, Interferon-gamma immunology, Lipid Peroxidation, Melanoma genetics, Melanoma immunology, Melanoma metabolism, Melanoma therapy, Mice, Neoplasms metabolism, Nivolumab therapeutic use, Reactive Oxygen Species metabolism, Treatment Outcome, CD8-Positive T-Lymphocytes immunology, Ferroptosis drug effects, Immunotherapy, Neoplasms immunology, Neoplasms therapy
- Abstract
Cancer immunotherapy restores or enhances the effector function of CD8
+ T cells in the tumour microenvironment1,2 . CD8+ T cells activated by cancer immunotherapy clear tumours mainly by inducing cell death through perforin-granzyme and Fas-Fas ligand pathways3,4 . Ferroptosis is a form of cell death that differs from apoptosis and results from iron-dependent accumulation of lipid peroxide5,6 . Although it has been investigated in vitro7,8 , there is emerging evidence that ferroptosis might be implicated in a variety of pathological scenarios9,10 . It is unclear whether, and how, ferroptosis is involved in T cell immunity and cancer immunotherapy. Here we show that immunotherapy-activated CD8+ T cells enhance ferroptosis-specific lipid peroxidation in tumour cells, and that increased ferroptosis contributes to the anti-tumour efficacy of immunotherapy. Mechanistically, interferon gamma (IFNγ) released from CD8+ T cells downregulates the expression of SLC3A2 and SLC7A11, two subunits of the glutamate-cystine antiporter system xc - , impairs the uptake of cystine by tumour cells, and as a consequence, promotes tumour cell lipid peroxidation and ferroptosis. In mouse models, depletion of cystine or cysteine by cyst(e)inase (an engineered enzyme that degrades both cystine and cysteine) in combination with checkpoint blockade synergistically enhanced T cell-mediated anti-tumour immunity and induced ferroptosis in tumour cells. Expression of system xc - was negatively associated, in cancer patients, with CD8+ T cell signature, IFNγ expression, and patient outcome. Analyses of human transcriptomes before and during nivolumab therapy revealed that clinical benefits correlate with reduced expression of SLC3A2 and increased IFNγ and CD8. Thus, T cell-promoted tumour ferroptosis is an anti-tumour mechanism, and targeting this pathway in combination with checkpoint blockade is a potential therapeutic approach.- Published
- 2019
- Full Text
- View/download PDF
40. Spatial and phenotypic immune profiling of metastatic colon cancer.
- Author
-
Lazarus J, Maj T, Smith JJ, Perusina Lanfranca M, Rao A, D'Angelica MI, Delrosario L, Girgis A, Schukow C, Shia J, Kryczek I, Shi J, Wasserman I, Crawford H, Nathan H, Pasca Di Magliano M, Zou W, and Frankel TL
- Subjects
- B7-H1 Antigen metabolism, Cancer Survivors, Colonic Neoplasms genetics, Colonic Neoplasms pathology, DNA Mismatch Repair, Humans, Immunohistochemistry, Immunophenotyping, Microsatellite Repeats, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Regulatory immunology, Tumor Microenvironment, Colonic Neoplasms immunology, Liver Neoplasms secondary
- Abstract
Paramount to the efficacy of immune checkpoint inhibitors is proper selection of patients with adequate tumor immunogenicity and a robust but suppressed immune infiltrate. In colon cancer, immune-based therapies are approved for patients with DNA mismatch repair (MMR) deficiencies, in whom accumulation of genetic mutations results in increased neoantigen expression, triggering an immune response that is suppressed by the PD-L1/PD-1 pathway. Here, we report that characterization of the microenvironment of MMR-deficient metastatic colorectal cancer using multiplex fluorescent immunohistochemistry (mfIHC) identified increased infiltration of cytotoxic T lymphocytes (CTLs), which were more often engaged with epithelial cells (ECs) and improved overall survival. A subset of patients with intact MMR but a similar immune microenvironment to MMR-deficient patients was identified and found to universally express high levels of PD-L1, suggesting that they may represent a currently untreated, checkpoint inhibitor-responsive population. Further, PD-L1 expression on antigen-presenting cells (APCs) in the tumor microenvironment (TME) resulted in impaired CTL/EC engagement and enhanced infiltration and engagement of Tregs. Characterization of the TME by mfIHC highlights the interconnection between immunity and immunosuppression in metastatic colon cancer and may better stratify patients for receipt of immunotherapies.
- Published
- 2018
- Full Text
- View/download PDF
41. Human Naive T Cells Express Functional CXCL8 and Promote Tumorigenesis.
- Author
-
Crespo J, Wu K, Li W, Kryczek I, Maj T, Vatan L, Wei S, Opipari AW, and Zou W
- Subjects
- Animals, Carcinogenesis, Cell Line, Tumor, Cell Movement, Cytokines metabolism, Gene Expression Regulation, Humans, Interleukin-8 genetics, Lymphocyte Activation, Mice, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Xenograft Model Antitumor Assays, Blood Cells physiology, Interleukin-8 metabolism, Neoplasms, Experimental immunology, Neutrophils physiology, T-Lymphocytes physiology, Umbilical Cord pathology
- Abstract
Naive T cells are thought to be functionally quiescent. In this study, we studied and compared the phenotype, cytokine profile, and potential function of human naive CD4
+ T cells in umbilical cord and peripheral blood. We found that naive CD4+ T cells, but not memory T cells, expressed high levels of chemokine CXCL8. CXCL8+ naive T cells were preferentially enriched CD31+ T cells and did not express T cell activation markers or typical Th effector cytokines, including IFN-γ, IL-4, IL-17, and IL-22. In addition, upon activation, naive T cells retained high levels of CXCL8 expression. Furthermore, we showed that naive T cell-derived CXCL8 mediated neutrophil migration in the in vitro migration assay, supported tumor sphere formation, and promoted tumor growth in an in vivo human xenograft model. Thus, human naive T cells are phenotypically and functionally heterogeneous and can carry out active functions in immune responses., (Copyright © 2018 by The American Association of Immunologists, Inc.)- Published
- 2018
- Full Text
- View/download PDF
42. Aerobic Glycolysis Controls Myeloid-Derived Suppressor Cells and Tumor Immunity via a Specific CEBPB Isoform in Triple-Negative Breast Cancer.
- Author
-
Li W, Tanikawa T, Kryczek I, Xia H, Li G, Wu K, Wei S, Zhao L, Vatan L, Wen B, Shu P, Sun D, Kleer C, Wicha M, Sabel M, Tao K, Wang G, and Zou W
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Female, Humans, Mice, CCAAT-Enhancer-Binding Protein-beta metabolism, Glycolysis, Granulocyte Colony-Stimulating Factor metabolism, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Immune Tolerance, Myeloid-Derived Suppressor Cells immunology, Triple Negative Breast Neoplasms immunology
- Abstract
Myeloid-derived suppressor cells (MDSCs) inhibit anti-tumor immunity. Aerobic glycolysis is a hallmark of cancer. However, the link between MDSCs and glycolysis is unknown in patients with triple-negative breast cancer (TNBC). Here, we detect abundant glycolytic activities in human TNBC. In two TNBC mouse models, 4T1 and Py8119, glycolysis restriction inhibits tumor granulocyte colony-stimulating factor (G-CSF) and granulocyte macrophage colony-stimulating factor (GM-CSF) expression and reduces MDSCs. These are accompanied with enhanced T cell immunity, reduced tumor growth and metastasis, and prolonged mouse survival. Mechanistically, glycolysis restriction represses the expression of a specific CCAAT/enhancer-binding protein beta (CEBPB) isoform, liver-enriched activator protein (LAP), via the AMP-activated protein kinase (AMPK)-ULK1 and autophagy pathways, whereas LAP controls G-CSF and GM-CSF expression to support MDSC development. Glycolytic signatures that include lactate dehydrogenase A correlate with high MDSCs and low T cells, and are associated with poor human TNBC outcome. Collectively, tumor glycolysis orchestrates a molecular network of the AMPK-ULK1, autophagy, and CEBPB pathways to affect MDSCs and maintain tumor immunosuppression., (Published by Elsevier Inc.)
- Published
- 2018
- Full Text
- View/download PDF
43. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression.
- Author
-
Lin H, Wei S, Hurt EM, Green MD, Zhao L, Vatan L, Szeliga W, Herbst R, Harms PW, Fecher LA, Vats P, Chinnaiyan AM, Lao CD, Lawrence TS, Wicha M, Hamanishi J, Mandai M, Kryczek I, and Zou W
- Published
- 2018
- Full Text
- View/download PDF
44. miR-508 Defines the Stem-like/Mesenchymal Subtype in Colorectal Cancer.
- Author
-
Yan TT, Ren LL, Shen CQ, Wang ZH, Yu YN, Liang Q, Tang JY, Chen YX, Sun DF, Zgodzinski W, Majewski M, Radwan P, Kryczek I, Zhong M, Chen J, Liu Q, Zou W, Chen HY, Hong J, and Fang JY
- Subjects
- Animals, Antigens, CD biosynthesis, Caco-2 Cells, Cadherins biosynthesis, Cell Line, Tumor, Cell Movement genetics, HCT116 Cells, HT29 Cells, Humans, Lung Neoplasms secondary, Mice, Mice, Inbred BALB C, Mice, Inbred NOD, Mice, Nude, Mice, SCID, Neoplasm Invasiveness genetics, Neoplasm Transplantation, Polycomb Repressive Complex 1 biosynthesis, RNA, Long Noncoding biosynthesis, RNA, Long Noncoding genetics, Transcription Factors biosynthesis, Transplantation, Heterologous, Zinc Finger E-box-Binding Homeobox 1 biosynthesis, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Epithelial-Mesenchymal Transition genetics, Mesenchymal Stem Cells cytology, MicroRNAs genetics
- Abstract
Colorectal cancer includes an invasive stem-like/mesenchymal subtype, but its genetic drivers, functional, and clinical relevance are uncharacterized. Here we report the definition of an altered miRNA signature defining this subtype that includes a major genomic loss of miR-508. Mechanistic investigations showed that this miRNA affected the expression of cadherin CDH1 and the transcription factors ZEB1, SALL4, and BMI1. Loss of miR-508 in colorectal cancer was associated with upregulation of the novel hypoxia-induced long noncoding RNA AK000053. Ectopic expression of miR-508 in colorectal cancer cells blunted epithelial-to-mesenchymal transition (EMT), stemness, migration, and invasive capacity in vitro and in vivo In clinical colorectal cancer specimens, expression of miR-508 negatively correlated with stemness and EMT-associated gene expression and positively correlated with patient survival. Overall, our results showed that miR-508 is a key functional determinant of the stem-like/mesenchymal colorectal cancer subtype and a candidate therapeutic target for its treatment. Significance: These results define a key functional determinant of a stem-like/mesenchymal subtype of colorectal cancers and a candidate therapeutic target for its treatment. Cancer Res; 78(7); 1751-65. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
45. High expression of CC chemokine receptor 5 (CCR5) promotes disease progression in patients with B-cell non-Hodgkin lymphomas.
- Author
-
Butrym A, Kryczek I, Dlubek D, Jaskula E, Lange A, Jurczyszyn A, and Mazur G
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Tumor genetics, Disease Progression, Female, Gene Expression Regulation, Neoplastic, Humans, Lymphoma, B-Cell pathology, Lymphoma, Non-Hodgkin pathology, Male, Middle Aged, Prognosis, Lymphoma, B-Cell genetics, Lymphoma, Non-Hodgkin genetics, Receptors, CCR5 genetics
- Abstract
Chemokines are small proteins, that regulate cell migration in many physiological and pathologic processes in human body. They are also responsible for cancer progression. CC chemokine receptor 5 (CCR5) is responsible for cell recruitment in inflammation and may be involved in antitumor immune response controlling. Aberrant CCR5 can be found in different kind of cancers, not only hematological, but also solid tumors. Non-Hodgkin lymphomas consist of many lymphoma subtypes. They predominantly derive from B cells and can have very heterogenous clinical course. That is why new prognostic factors are still needed to predict and select high-risk patients. We evaluated CCR5 expression in lymph nodes derived from B-cell lymphomas in comparison to reactive lymphatic tissue (reactive lymph nodes): samples of lymphoma lymph nodes, peripheral blood, and bone marrow aspirates of patients with B-cell non-Hodgkin lymphoma were taken at diagnosis and after completed chemotherapy. Gene expression was determined by the reverse transcription-polymerase chain reaction method. Expression was estimated from 0AU (no amplificate signal) to 3AU (maximal amplificate signal). We found low CCR5 expression in lymphomas and reactive lymph nodes. Higher CCR5 gene expression in lymphoma patients was correlated with advanced stage of the disease, high proliferation index (Ki-67), and international prognostic index. Patients with higher CCR5 expression had shorter survival. CCR5 high expression may have a role in non-Hodgkin's lymphomas progression and can influence patients' survival. CCR5 also can become an immunotherapeutic target for novel treatment options in the future as well as new prognostic factor., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
46. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor.
- Author
-
Maj T, Wang W, Crespo J, Zhang H, Wang W, Wei S, Zhao L, Vatan L, Shao I, Szeliga W, Lyssiotis C, Liu JR, Kryczek I, and Zou W
- Subjects
- 5'-Nucleotidase genetics, 5'-Nucleotidase metabolism, Adenosine metabolism, Animals, Antigens, CD metabolism, Apyrase metabolism, CTLA-4 Antigen metabolism, Female, GPI-Linked Proteins genetics, Humans, Interleukin-10 metabolism, Interleukins metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, NF-E2-Related Factor 2 metabolism, Oxygen metabolism, Receptor, Adenosine A2A metabolism, Transforming Growth Factor beta metabolism, Tumor Cells, Cultured, Tumor Microenvironment immunology, Apoptosis immunology, B7-H1 Antigen metabolism, Immune Tolerance immunology, Ovarian Neoplasms immunology, Oxidative Stress physiology, T-Lymphocytes, Regulatory immunology
- Abstract
Live regulatory T cells (T
reg cells) suppress antitumor immunity, but how Treg cells behave in the metabolically abnormal tumor microenvironment remains unknown. Here we show that tumor Treg cells undergo apoptosis, and such apoptotic Treg cells abolish spontaneous and PD-L1-blockade-mediated antitumor T cell immunity. Biochemical and functional analyses show that adenosine, but not typical suppressive factors such as PD-L1, CTLA-4, TGF-β, IL-35, and IL-10, contributes to apoptotic Treg -cell-mediated immunosuppression. Mechanistically, apoptotic Treg cells release and convert a large amount of ATP to adenosine via CD39 and CD73, and mediate immunosuppression via the adenosine and A2A pathways. Apoptosis in Treg cells is attributed to their weak NRF2-associated antioxidant system and high vulnerability to free oxygen species in the tumor microenvironment. Thus, the data support a model wherein tumor Treg cells sustain and amplify their suppressor capacity through inadvertent death via oxidative stress. This work highlights the oxidative pathway as a metabolic checkpoint that controls Treg cell behavior and affects the efficacy of therapeutics targeting cancer checkpoints.- Published
- 2017
- Full Text
- View/download PDF
47. Suppression of FIP200 and autophagy by tumor-derived lactate promotes naïve T cell apoptosis and affects tumor immunity.
- Author
-
Xia H, Wang W, Crespo J, Kryczek I, Li W, Wei S, Bian Z, Maj T, He M, Liu RJ, He Y, Rattan R, Munkarah A, Guan JL, and Zou W
- Subjects
- Animals, Apoptosis genetics, Autophagy genetics, Autophagy-Related Proteins, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Lactic Acid metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Neoplasms, Experimental genetics, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Protein-Tyrosine Kinases genetics, T-Lymphocytes metabolism, Apoptosis drug effects, Autophagy drug effects, Lactic Acid pharmacology, Ovarian Neoplasms metabolism, Protein-Tyrosine Kinases metabolism, T-Lymphocytes drug effects
- Abstract
Naïve T cells are poorly studied in cancer patients. We report that naïve T cells are prone to undergo apoptosis due to a selective loss of FAK family-interacting protein of 200 kDa (FIP200) in ovarian cancer patients and tumor-bearing mice. This results in poor antitumor immunity via autophagy deficiency, mitochondria overactivation, and high reactive oxygen species production in T cells. Mechanistically, loss of FIP200 disables the balance between proapoptotic and antiapoptotic Bcl-2 family members via enhanced argonaute 2 (Ago2) degradation, reduced Ago2 and microRNA1198-5p complex formation, less microRNA1198-5p maturation, and consequently abolished microRNA1198-5p-mediated repression on apoptotic gene Bak1 Bcl-2 overexpression and mitochondria complex I inhibition rescue T cell apoptosis and promoted tumor immunity. Tumor-derived lactate translationally inhibits FIP200 expression by down-regulating the nicotinamide adenine dinucleotide level while potentially up-regulating the inhibitory effect of adenylate-uridylate-rich elements within the 3' untranslated region of Fip200 mRNA. Thus, tumors metabolically target naïve T cells to evade immunity., (Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2017
- Full Text
- View/download PDF
48. Phenotype and tissue distribution of CD28H + immune cell subsets.
- Author
-
Crespo J, Vatan L, Maj T, Liu R, Kryczek I, and Zou W
- Abstract
CD28H is a newly discovered co-receptor of the human B7 family. CD28H interacts with its ligand B7-H5 and regulates T cell response. Here we showed that CD28H was not expressed on granulocytes, monocytes, myeloid dendritic cells (MDCs), and B cells, but constitutively expressed with moderate levels on memory T cells and with high levels on naïve T cells, innate lymphoid cells (ILCs), natural killer (NK) cells, and plasmacytoid dendritic cells (PDCs) in human peripheral blood. Similar CD28H
+ cell profile existed in secondary lymphoid organs and pathological tissues including multiple types of cancers. Further analysis demonstrated that CD28H+ naïve and CD28H+ memory T cells were characterized with increased naïve feature and less effector functional phenotype, respectively. High levels of constitutive CD28H expression on naïve T cells and innate immune cells suggest a potential role of CD28H in innate and adaptive immunity.- Published
- 2017
- Full Text
- View/download PDF
49. Fusobacterium nucleatum Promotes Chemoresistance to Colorectal Cancer by Modulating Autophagy.
- Author
-
Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N, Chen Y, Chen H, Hong J, Zou W, and Fang JY
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Capecitabine therapeutic use, Colorectal Neoplasms metabolism, Drug Resistance, Neoplasm, Heterografts, Mice, MicroRNAs metabolism, Neoplasm Transplantation, Platinum Compounds therapeutic use, Recurrence, Toll-Like Receptors metabolism, Tumor Microenvironment, Autophagy, Colorectal Neoplasms drug therapy, Colorectal Neoplasms pathology, Fusobacterium nucleatum physiology, Gastrointestinal Microbiome
- Abstract
Gut microbiota are linked to chronic inflammation and carcinogenesis. Chemotherapy failure is the major cause of recurrence and poor prognosis in colorectal cancer patients. Here, we investigated the contribution of gut microbiota to chemoresistance in patients with colorectal cancer. We found that Fusobacterium (F.) nucleatum was abundant in colorectal cancer tissues in patients with recurrence post chemotherapy, and was associated with patient clinicopathological characterisitcs. Furthermore, our bioinformatic and functional studies demonstrated that F. nucleatum promoted colorectal cancer resistance to chemotherapy. Mechanistically, F. nucleatum targeted TLR4 and MYD88 innate immune signaling and specific microRNAs to activate the autophagy pathway and alter colorectal cancer chemotherapeutic response. Thus, F. nucleatum orchestrates a molecular network of the Toll-like receptor, microRNAs, and autophagy to clinically, biologically, and mechanistically control colorectal cancer chemoresistance. Measuring and targeting F. nucleatum and its associated pathway will yield valuable insight into clinical management and may ameliorate colorectal cancer patient outcomes., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
50. IL33 Promotes Colon Cancer Cell Stemness via JNK Activation and Macrophage Recruitment.
- Author
-
Fang M, Li Y, Huang K, Qi S, Zhang J, Zgodzinski W, Majewski M, Wallner G, Gozdz S, Macek P, Kowalik A, Pasiarski M, Grywalska E, Vatan L, Nagarsheth N, Li W, Zhao L, Kryczek I, Wang G, Wang Z, Zou W, and Wang L
- Subjects
- Animals, Cell Line, Tumor, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Chemotaxis, Leukocyte genetics, Chemotaxis, Leukocyte immunology, Colonic Neoplasms immunology, Colonic Neoplasms mortality, Disease Models, Animal, Gene Expression, Heterografts, Humans, Interleukin-1 Receptor-Like 1 Protein genetics, Interleukin-1 Receptor-Like 1 Protein metabolism, Macrophages immunology, Male, Mice, Prognosis, Proto-Oncogene Proteins c-jun genetics, Proto-Oncogene Proteins c-jun metabolism, Tumor Cells, Cultured, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Interleukin-33 metabolism, JNK Mitogen-Activated Protein Kinases metabolism, Macrophages metabolism, Neoplastic Stem Cells metabolism
- Abstract
The expression and biological role of IL33 in colon cancer is poorly understood. In this study, we show that IL33 is expressed by vascular endothelial cells and tumor cells in the human colon cancer microenvironment. Administration of human IL33 and overexpression of murine IL33 enhanced human and murine colon cancer cell growth in vivo , respectively. IL33 stimulated cell sphere formation and prevented chemotherapy-induced tumor apoptosis. Mechanistically, IL33 activated core stem cell genes NANOG, NOTCH3 , and OCT3 / 4 via the ST2 signaling pathway, and induced phosphorylation of c-Jun N terminal kinase (JNK) activation and enhanced binding of c-Jun to the promoters of the core stem cell genes. Moreover, IL33 recruited macrophages into the cancer microenvironment and stimulated them to produce prostaglandin E
2 , which supported colon cancer stemness and tumor growth. Clinically, tumor IL33 expression associated with poor survival in patients with metastatic colon cancer. Thus, IL33 dually targets tumor cells and macrophages and endows stem-like qualities to colon cancer cells to promote carcinogenesis. Collectively, our work reveals an immune-associated mechanism that extrinsically confers cancer cell stemness properties. Targeting the IL33 signaling pathway may offer an opportunity to treat patients with metastatic cancer. Cancer Res; 77(10); 2735-45. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.