Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Marcin Kortylewski is active.

Publication


Featured researches published by Marcin Kortylewski.


Nature Reviews Immunology | 2007

Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment

Hua Yu; Marcin Kortylewski; Drew M. Pardoll

Immune cells in the tumour microenvironment not only fail to mount an effective anti-tumour immune response, but also interact intimately with the transformed cells to promote oncogenesis actively. Signal transducer and activator of transcription 3 (STAT3), which is a point of convergence for numerous oncogenic signalling pathways, is constitutively activated both in tumour cells and in immune cells in the tumour microenvironment. Constitutively activated STAT3 inhibits the expression of mediators necessary for immune activation against tumour cells. Furthermore, STAT3 activity promotes the production of immunosuppressive factors that activate STAT3 in diverse immune-cell subsets, altering gene-expression programmes and, thereby, restraining anti-tumour immune responses. As such, STAT3 propagates several levels of crosstalk between tumour cells and their immunological microenvironment, leading to tumour-induced immunosuppression. Consequently, STAT3 has emerged as a promising target for cancer immunotherapy.


Nature Medicine | 2005

Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity

Marcin Kortylewski; Maciej Kujawski; Tianhong Wang; Sheng Wei; Shumin M. Zhang; Shari Pilon-Thomas; Guilian Niu; Heidi Kay; James J. Mulé; William G. Kerr; Richard Jove; Drew M. Pardoll; Hua Yu

The immune system can act as an extrinsic suppressor of tumors. Therefore, tumor progression depends in part on mechanisms that downmodulate intrinsic immune surveillance. Identifying these inhibitory pathways may provide promising targets to enhance antitumor immunity. Here, we show that Stat3 is constitutively activated in diverse tumor-infiltrating immune cells, and ablating Stat3 in hematopoietic cells triggers an intrinsic immune-surveillance system that inhibits tumor growth and metastasis. We observed a markedly enhanced function of dendritic cells, T cells, natural killer (NK) cells and neutrophils in tumor-bearing mice with Stat3−/− hematopoietic cells, and showed that tumor regression requires immune cells. Targeting Stat3 with a small-molecule drug induces T cell– and NK cell–dependent growth inhibition of established tumors otherwise resistant to direct killing by the inhibitor. Our findings show that Stat3 signaling restrains natural tumor immune surveillance and that inhibiting hematopoietic Stat3 in tumor-bearing hosts elicits multicomponent therapeutic antitumor immunity.


Nature Reviews Immunology | 2007

Tumour immunology: Crosstalk between cancer and immune cells: role of STAT3 in the tumour microenvironment

Hua Yu; Marcin Kortylewski; Drew M. Pardoll

Immune cells in the tumour microenvironment not only fail to mount an effective anti-tumour immune response, but also interact intimately with the transformed cells to promote oncogenesis actively. Signal transducer and activator of transcription 3 (STAT3), which is a point of convergence for numerous oncogenic signalling pathways, is constitutively activated both in tumour cells and in immune cells in the tumour microenvironment. Constitutively activated STAT3 inhibits the expression of mediators necessary for immune activation against tumour cells. Furthermore, STAT3 activity promotes the production of immunosuppressive factors that activate STAT3 in diverse immune-cell subsets, altering gene-expression programmes and, thereby, restraining anti-tumour immune responses. As such, STAT3 propagates several levels of crosstalk between tumour cells and their immunological microenvironment, leading to tumour-induced immunosuppression. Consequently, STAT3 has emerged as a promising target for cancer immunotherapy.


Journal of Experimental Medicine | 2009

IL-17 can promote tumor growth through an IL-6–Stat3 signaling pathway

Lin Wang; Tangsheng Yi; Marcin Kortylewski; Drew M. Pardoll; Defu Zeng; Hua Yu

Although the Th17 subset and its signature cytokine, interleukin (IL)-17A (IL-17), are implicated in certain autoimmune diseases, their role in cancer remains to be further explored. IL-17 has been shown to be elevated in several types of cancer, but how it might contribute to tumor growth is still unclear. We show that growth of B16 melanoma and MB49 bladder carcinoma is reduced in IL-17−/− mice but drastically accelerated in IFN-γ−/− mice, contributed to by elevated intratumoral IL-17, indicating a role of IL-17 in promoting tumor growth. Adoptive transfer studies and analysis of the tumor microenvironment suggest that CD4+ T cells are the predominant source of IL-17. Enhancement of tumor growth by IL-17 involves direct effects on tumor cells and tumor-associated stromal cells, which bear IL-17 receptors. IL-17 induces IL-6 production, which in turn activates oncogenic signal transducer and activator of transcription (Stat) 3, up-regulating prosurvival and proangiogenic genes. The Th17 response can thus promote tumor growth, in part via an IL-6–Stat3 pathway.


Journal of Immunology | 2007

Cutting Edge: An In Vivo Requirement for STAT3 Signaling in TH17 Development and TH17-Dependent Autoimmunity

Timothy J. Harris; Joseph F. Grosso; Hung-Rong Yen; Hong Xin; Marcin Kortylewski; Emilia Albesiano; Edward L. Hipkiss; Derese Getnet; Monica V. Goldberg; Charles H. Maris; Franck Housseau; Hua Yu; Drew M. Pardoll; Charles G. Drake

STAT3 activation has been observed in several autoimmune diseases, suggesting that STAT3-mediated pathways promote pathologic immune responses. We provide in vivo evidence that the fundamental role of STAT3 signaling in autoimmunity relates to its absolute requirement for generating TH17 T cell responses. We show that STAT3 is a master regulator of this pathogenic T cell subtype, acting at multiple levels in vivo, including TH17 T cell differentiation and cytokine production, as well as induction of RORγt and the IL-23R. Neither naturally occurring TH17 cells nor TH17-dependent autoimmunity occurs when STAT3 is ablated in CD4 cells. Furthermore, ablation of STAT3 signaling in CD4 cells results in increased TH1 responses, indicating that STAT3 signaling skews TH responses away from the TH1 pathway and toward the TH17 pathway. Thus, STAT3 is a candidate target for TH17-dependent autoimmune disease immunotherapy that could selectively inhibit pathogenic immune pathways.


Cancer Cell | 2009

Regulation of the IL-23 and IL-12 balance by Stat3 signaling in the tumor microenvironment.

Marcin Kortylewski; Hong Xin; Maciej Kujawski; Heehyoung Lee; Yong Liu; Timothy J. Harris; Charles G. Drake; Drew M. Pardoll; Hua Yu

Interactions between tumor and immune cells either enhance or inhibit cancer progression. We show here that Stat3 signaling within the tumor microenvironment induces a procarcinogenic cytokine, IL-23, while inhibiting a central anticarcinogenic cytokine, IL-12, thereby shifting the balance of tumor immunity toward carcinogenesis. Stat3 induces expression of IL-23, which is mainly produced by tumor-associated macrophages, via direct transcriptional activation of the IL-23/p19 gene. Furthermore, Stat3 inhibits NF-kappaB/c-Rel-dependent IL-12/p35 gene expression in tumor-associated dendritic cells. Tumor-associated regulatory T cells (Tregs) express IL-23 receptor, which activates Stat3 in this cell type, leading to upregulation of the Treg-specific transcription factor Foxp3 and the immunosuppressive cytokine IL-10. These results demonstrate that Stat3 promotes IL-23-mediated procarcinogenic immune responses while inhibiting IL-12-dependent antitumor immunity.


Nature Biotechnology | 2009

In vivo delivery of siRNA to immune cells by conjugation to a TLR9 agonist enhances antitumor immune responses

Marcin Kortylewski; Piotr Swiderski; Andreas Herrmann; Lin Wang; Claudia M. Kowolik; Maciej Kujawski; Heehyoung Lee; Anna Scuto; Yong Liu; Chunmei Yang; Jiehui Deng; Harris S. Soifer; Andrew Raubitschek; Stephen J. Forman; John J. Rossi; Drew M. Pardoll; Richard Jove; Hua Yu

Efficient delivery of small interfering (si)RNA to specific cell populations in vivo remains a formidable challenge to its successful therapeutic application. We show that siRNA synthetically linked to a CpG oligonucleotide agonist of toll-like receptor (TLR)9 targets and silences genes in TLR9+ myeloid cells and B cells, both of which are key components of the tumor microenvironment. When a CpG-conjugated siRNA that targets the immune suppressor gene Stat3 is injected in mice either locally at the tumor site or intravenously, it enters tumor-associated dendritic cells, macrophages and B cells. Silencing of Stat3 leads to activation of tumor-associated immune cells and ultimately to potent antitumor immune responses. Our findings demonstrate the potential of TLR agonist–siRNA conjugates for targeted gene silencing coupled with TLR stimulation and immune activation in the tumor microenvironment.Efficient delivery of siRNA to specific cell populations in vivo remains a formidable challenge to its successful therapeutic application. We describe a novel siRNA-based approach – synthetically linking siRNA to an oligonucleotide TLR9 agonist – that targets and silences genes in TLR9+ myeloid cells and B cells, both of which are key components of the tumor microenvironment. Because Stat3 in tumor-associated immune cells suppresses antitumor immune responses and hinders TLR9-induced immune stimulation, we tested CpG-Stat3siRNA conjugates for anti-tumor effects. When injected locally at the tumor site or systemically through an intravenous route, the CpG-Stat3siRNA conjugates access tumor-associated dendritic cells, macrophages and B cells, inhibit Stat3 expression, leading to activation of tumor-associated immune cells, and ultimately potent anti-tumor immune responses. Our findings demonstrate the potential of TLR agonist-siRNA conjugates for targeted gene silencing coupled with TLR stimulation and immune activation in the tumor microenvironment.


Current Opinion in Immunology | 2008

Role of Stat3 in suppressing anti-tumor immunity.

Marcin Kortylewski; Hua Yu

Recent evidence suggests that what sustains a tumors survival and proliferation can also subvert the immune system from its defense role. As a point of convergence for numerous oncogenic signaling pathways, Stat3 is constitutively activated in diverse human cancer cells. Activated Stat3 not only upregulates genes crucial for survival, proliferation, angiogenesis, and metastasis but also promotes expression of immune suppressive factors while inhibiting Th1 immunostimulatory molecules. By virtue of its ability to promote expression of many factors that activate Stat3 in diverse cells, Stat3 allows malignant and immune cells resonate, forming close partnership for tumor immune evasion, tumor progression, and resistance to therapies.


Glia | 2009

Stat3 inhibition activates tumor macrophages and abrogates glioma growth in mice

Leying Zhang; Darya Alizadeh; Michelle Van Handel; Marcin Kortylewski; Hua Yu; Behnam Badie

As the main effector‐cell population of the central nervous system, microglia (MG) are considered to play an important immunoregulatory function in a number of pathological conditions such as inflammation, trauma, degenerative disease, and brain tumors. Recent studies, however, have suggested that the anti‐neoplastic function of MG may be suppressed in malignant brain tumors. Considering the proposed suppressive role of signal transducers and activators of transcription 3 (Stat3) in antitumor immunity, we evaluated the role of Stat3 inhibition on MG and macrophage (MP) activation and tumor growth in a murine glioma model. N9 MG cells were exposed to GL261 glioma conditioned medium (GL261‐CM) and evaluated for Stat3 activity and cytokine expression. Furthermore, the role of Stat3 inhibition on MG and MP activation was studied both in vitro and in vivo. Finally, the effect of Stat3 inhibition on tumor growth was assessed in intracranial GL261 gliomas. GL261‐CM increased Stat3 activity in N9 cells in vitro and resulted in overexpression of IL‐10 and IL‐6, and downregulation of IL1‐β, a pro‐inflammatory cytokine. Inhibition of Stat3 by CPA‐7 or siRNA reversed glioma‐induced cytokine expression profile in N9 cells. Furthermore, inactivation of Stat3 in intracranial GL261 tumors by siRNA resulted in MG/MP activation and tumor growth inhibition. Glioma‐induced MG and MP suppression may be mediated thorough Stat3. Inhibition of Stat3 function in tumor MG/MP may result in their activation and can potentially be used as an adjunct immunotherapy approach for gliomas.


Scientific Reports | 2015

Macrophage immunomodulation by breast cancer-derived exosomes requires Toll-like receptor 2-mediated activation of NF-κB

Amy Chow; Weiying Zhou; Liang Liu; Miranda Y. Fong; Jackson Champer; Desiree Van Haute; Andrew R. Chin; Xiubao Ren; Bogdan Gabriel Gugiu; Zhipeng Meng; Wendong Huang; Vu N. Ngo; Marcin Kortylewski; Shizhen Emily Wang

Growing evidence links tumor progression with chronic inflammatory processes and dysregulated activity of various immune cells. In this study, we demonstrate that various types of macrophages internalize microvesicles, called exosomes, secreted by breast cancer and non-cancerous cell lines. Although both types of exosomes targeted macrophages, only cancer-derived exosomes stimulated NF-κB activation in macrophages resulting in secretion of pro-inflammatory cytokines such as IL-6, TNFα, GCSF, and CCL2. In vivo mouse experiments confirmed that intravenously injected exosomes are efficiently internalized by macrophages in the lung and brain, which correlated with upregulation of inflammatory cytokines. In mice bearing xenografted human breast cancers, tumor-derived exosomes were internalized by macrophages in axillary lymph nodes thereby triggering expression of IL-6. Genetic ablation of Toll-like receptor 2 (TLR2) or MyD88, a critical signaling adaptor in the NF-κB pathway, completely abolished the effect of tumor-derived exosomes. In contrast, inhibition of TLR4 or endosomal TLRs (TLR3/7/8/9) failed to abrogate NF-κB activation by exosomes. We further found that palmitoylated proteins present on the surface of tumor-secreted exosomes contributed to NF-κB activation. Thus, our results highlight a novel mechanism used by breast cancer cells to induce pro-inflammatory activity of distant macrophages through circulating exosomal vesicles secreted during cancer progression.

Collaboration


Dive into the Marcin Kortylewski's collaboration.

Top Co-Authors

Avatar

Hua Yu

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sumanta K. Pal

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Piotr Swiderski

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dayson Moreira

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stephen J. Forman

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jeremy O. Jones

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Qifang Zhang

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dewan Md Sakib Hossain

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Claudia M. Kowolik

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sergey Nechaev

City of Hope National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge