Network


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

Hotspot


Dive into the research topics where Kristina Young is active.

Publication


Featured researches published by Kristina Young.


Radiation Research | 2014

Expression of Arginase I in Myeloid Cells Limits Control of Residual Disease after Radiation Therapy of Tumors in Mice

Marka Crittenden; Talicia Savage; Benjamin Cottam; Jason R. Baird; Paulo C. Rodriguez; Pippa Newell; Kristina Young; Andrew M. Jackson; Michael J. Gough

An accumulating body of evidence demonstrates that radiation therapy can generate adaptive immune responses that contribute to tumor control. However, in the absence of additional immune therapy, the adaptive immune response is insufficient to prevent tumor recurrence or affect distant disease. It has been shown in multiple models that tumor-infiltrating myeloid cells exhibit alternative activation phenotypes and are able to suppress adaptive immune responses, and recent data suggests that the myeloid response in tumors treated with cytotoxic therapy limits tumor control. We hypothesized that tumor myeloid cells inhibit the adaptive immune response after radiation therapy through expression of the enzyme arginase I. Using a myeloid cell-specific deletion of arginase I in mice, we demonstrate an improved tumor control after radiation therapy. However, tumors still recurred despite the conditional knockdown of arginase I. Since multiple alternative factors may combine to inhibit adaptive immunity, we propose that targeting macrophage differentiation may be a more effective strategy than targeting individual suppressive pathways.


Cancer immunology research | 2014

TGFβ inhibition prior to hypofractionated radiation enhances efficacy in preclinical models

Kristina Young; Pippa Newell; Benjamin Cottam; David Friedman; Talicia Savage; Jason R. Baird; Emmanuel T. Akporiaye; Michael J. Gough; Marka Crittenden

Young and colleagues demonstrate in syngeneic mouse models of colorectal and pancreatic cancers that TGFβ inhibition with the oral, small-molecule inhibitor SM16 enhanced adaptive immunity in the tumor microenvironments and significantly improved the efficacy of subsequent radiotherapy. The immune infiltrate in colorectal cancer has been correlated with outcome, such that individuals with higher infiltrations of T cells have increased survival independent of the disease stage. For patients with lower immune infiltrates, overall survival is limited. Because the patients with colorectal cancer studied have received conventional cancer therapies, these data may indicate that the pretreatment tumor environment increases the efficacy of treatments such as chemotherapy, surgery, and radiotherapy. This study was designed to test the hypothesis that an improved immune environment in the tumor at the time of treatment will increase the efficacy of radiotherapy. We demonstrate that inhibition of TGFβ using the orally available small-molecule inhibitor SM16 improved the immune environment of tumors in mice and significantly improved the efficacy of subsequent radiotherapy. This effect was not due to changes in radiosensitivity, epithelial–mesenchymal transition, or changes in vascular function in the tumor; rather, this effect was dependent on adaptive immunity and resulted in long-term protective immunity in cured mice. These data demonstrate that immunotherapy is an option to improve the immune status of patients with poor tumor infiltrates and that pretreatment improves the efficacy of radiotherapy. Cancer Immunol Res; 2(10); 1011–22. ©2014 AACR.


Clinical & Developmental Immunology | 2013

The impact of the myeloid response to radiation therapy.

Michael J. Gough; Kristina Young; Marka Crittenden

Radiation therapy is showing potential as a partner for immunotherapies in preclinical cancer models and early clinical studies. As has been discussed elsewhere, radiation provides debulking, antigen and adjuvant release, and inflammatory targeting of effector cells to the treatment site, thereby assisting multiple critical checkpoints in antitumor adaptive immunity. Adaptive immunity is terminated by inflammatory resolution, an active process which ensures that inflammatory damage is repaired and tissue function is restored. We discuss how radiation therapy similarly triggers inflammation followed by repair, the consequences to adaptive immune responses in the treatment site, and how the myeloid response to radiation may impact immunotherapies designed to improve control of residual cancer cells.


Oncotarget | 2016

Mertk on tumor macrophages is a therapeutic target to prevent tumor recurrence following radiation therapy

Marka Crittenden; Jason R. Baird; David J. Friedman; Talicia Savage; Lauren Uhde; Alejandro Alice; Benjamin Cottam; Kristina Young; Pippa Newell; Cynthia Nguyen; Shelly Bambina; Gwen Kramer; Emmanuel T. Akporiaye; Anna Malecka; Andrew M. Jackson; Michael J. Gough

Radiation therapy provides a means to kill large numbers of cancer cells in a controlled location resulting in the release of tumor-specific antigens and endogenous adjuvants. However, by activating pathways involved in apoptotic cell recognition and phagocytosis, irradiated cancer cells engender suppressive phenotypes in macrophages. We demonstrate that the macrophage-specific phagocytic receptor, Mertk is upregulated in macrophages in the tumor following radiation therapy. Ligation of Mertk on macrophages results in anti-inflammatory cytokine responses via NF-kB p50 upregulation, which in turn limits tumor control following radiation therapy. We demonstrate that in immunogenic tumors, loss of Mertk is sufficient to permit tumor cure following radiation therapy. However, in poorly immunogenic tumors, TGFb inhibition is also required to result in tumor cure following radiation therapy. These data demonstrate that Mertk is a highly specific target whose absence permits tumor control in combination with radiation therapy.


Oncotarget | 2017

Neuroinflammatory and cognitive consequences of combined radiation and immunotherapy in a novel preclinical model

Gwendolyn J. McGinnis; David J. Friedman; Kristina Young; Eileen Ruth S. Torres; Charles R. Thomas; Michael J. Gough; Jacob Raber

Background Cancer patients often report behavioral and cognitive changes following cancer treatment. These effects can be seen in patients who have not yet received treatment or have received only peripheral (non-brain) irradiation. Novel treatments combining radiotherapy (RT) and immunotherapy (IT) demonstrate remarkable efficacy with respect to tumor outcomes by enhancing the proinflammatory environment in the tumor. However, a proinflammatory environment in the brain mediates cognitive impairments in other neurological disorders and may affect brain function in cancer patients receiving these novel treatments. Currently, gaps exist as to whether these treatments impact the brain in individuals with or without tumors and with regard to the underlying mechanisms. Results Combined treatment with precision RT and checkpoint inhibitor IT achieved control of tumor growth. However, BALB/c mice receiving combined treatment demonstrated changes in measures of anxiety levels, regardless of tumor status. C57BL/6J mice with tumors demonstrated increased anxiety, except following combined treatment. Object recognition memory was impaired in C57BL/6J mice without tumors following combined treatment. All mice with tumors showed impaired object recognition, except those treated with RT alone. Mice with tumors demonstrated impaired amygdala-dependent cued fear memory, while maintaining hippocampus-dependent context fear memory. These behavioral alterations and cognitive impairments were accompanied by increased microglial activation in mice receiving immunotherapy alone or combined with RT. Finally, based on tumor status, there were significant changes in proinflammatory cytokines (IFN-γ, IL-6, IL-5, IL-2, IL-10) and a growth factor (FGF-basic). Materials and Methods Here we test the hypothesis that IT combined with peripheral RT have detrimental behavioral and cognitive effects as a result of an enhanced proinflammatory environment in the brain. BALB/c mice with or without injected hind flank CT26 colorectal carcinoma or C57BL/6J mice with or without Lewis Lung carcinoma were used for all experiments. Checkpoint inhibitor IT, using an anti-CTLA-4 antibody, and precision CT-guided peripheral RT alone and combined were used to closely model clinical treatment. We assessed behavioral and cognitive performance and investigated the immune environment using immunohistochemistry and multiplex assays to analyze proinflammatory mediators. Conclusions Although combined treatment achieved tumor growth control, it affected the brain and induced changes in measures of anxiety, cognitive impairments, and neuroinflammation.


Hepatology Research | 2017

Programmed cell death-1 blockade enhances response to stereotactic radiation in an orthotopic murine model of hepatocellular carcinoma

David P. Friedman; Jason R. Baird; Kristina Young; Benjamin Cottam; Marka Crittenden; Scott L. Friedman; Michael J. Gough; Pippa Newell

Small, solitary hepatocellular carcinoma is curable with stereotactic radiation or other methods of tumor ablation, however, regional and systemic tumor recurrence occurs in over 70% of patients. Here we describe the ability of immunoradiotherapy to induce an antitumor immune response and delay the growth of tumors in immunocompetent mice.


Immunotherapy | 2015

Comparing equals when evaluating immunotherapy with different doses and fractions of radiation therapy.

Michael J. Gough; Marka Crittenden; Kristina Young

The combination of immunotherapy with radiation therapy is showing evidence of therapeutic synergy. Exciting preclinical data and the emergence of new clinically available immunotherapies has led to an explosion of combination therapies in the clinic. To understand the synergy of radiation and immunotherapy, increasing effort is going into investigating mechanisms of interaction in preclinical models. However, in one aspect we do not come close to modeling standard clinical scenarios. Few studies in mice recapitulate standard fractionation as given to patients. We discuss the problem in modeling clinically relevant biological equivalent dose in preclinical studies of immunotherapy and radiation therapy. As is well known in radiation oncology, repeated doses of radiation do not necessarily scale in a linear manner, such that 2 Gy delivered in five daily doses is not equivalent to 10 Gy in a single dose. In order to compare efficacy of different radiation regimens, an equation was developed to explain the biologic equivalent dose, BED, based on the in vivo survival curve. The BED equation is:


OncoImmunology | 2015

Tumor immune remodeling by TGFβ inhibition improves the efficacy of radiation therapy

Kristina Young; Michael J. Gough; Marka Crittenden

The tumor immune environment has been linked to prognosis in patients with a range of malignancies. Recently, we demonstrated in pre-clinical models that modifying the tumor immune environment using a small-molecule inhibitor of TGFb significantly improved outcome to subsequent radiation therapy. These data suggest that this and other immunotherapies may be used to remodel the tumor before conventional cancer therapies to improve outcomes.


Journal for ImmunoTherapy of Cancer | 2014

Preparative immunotherapy with anti-OX40 and anti-CTLA4 improves the response to chemotherapy

David Friedman; Kristina Young; Jason R. Baird; Benjamin Cottam; Talicia Savage; Pippa Newell; Melissa J. Kasiewicz; William L. Redmond; Brendan D. Curti; Todd Crocenzi; Michael J. Gough; Marka Crittenden

Meeting abstracts Recent studies have reported that decreased T cell infiltrate alone, or co-ordinate with increased macrophage infiltrate, correlate with decreased survival in a range of cancers, including patients with pancreatic cancer. Importantly, in mouse models of pancreatic cancer,


PLOS ONE | 2017

STING expression and response to treatment with STING ligands in premalignant and malignant disease

Jason R. Baird; Zipeng Feng; Hong D. Xiao; David P. Friedman; Ben Cottam; Bernard A. Fox; Gwen Kramer; Rom S. Leidner; R. Bryan Bell; Kristina Young; Marka Crittenden; Michael J. Gough

Human papilloma virus positive (HPV+) tumors represent a large proportion of anal, vulvar, vaginal, cervical and head and neck squamous carcinomas (HNSCC) and late stage invasive disease is thought to originate from a premalignant state. Cyclic dinucleotides that activate STimulator of INterferon Genes (STING) have been shown to cause rapid regression of a range of advanced tumors. We aimed to investigate STING ligands as a novel treatment for papilloma. We tested therapies in a spontaneous mouse model of papilloma of the face and anogenital region that histologically resembles human HPV-associated papilloma. We demonstrate that STING ligands cause rapid regression of papilloma, associated with T cell infiltration, and are significantly more effective than Imiquimod, a current immunotherapy for papilloma. In humans, we show that STING is expressed in the basal layer of normal skin and lost during keratinocyte differentiation. We found STING was expressed in all HPV-associated cervical and anal dysplasia and was strongly expressed in the cancer cells of HPV+ HNSCC but not in HPV-unrelated HNSCC. We found no strong association between STING expression and progressive disease in non-HPV oral dysplasia and oral pre-malignancies that are not HPV-related. These data demonstrate that STING is expressed in basal cells of the skin and is retained in HPV+ pre-malignancies and advanced cancers, but not in HPV-unrelated HNSCC. However, using a murine HNSCC model that does not express STING, we demonstrate that STING ligands are an effective therapy regardless of expression of STING by the cancer cells.

Collaboration


Dive into the Kristina Young's collaboration.

Top Co-Authors

Avatar

Michael J. Gough

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marka Crittenden

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar

Benjamin Cottam

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jason R. Baird

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar

Pippa Newell

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar

Talicia Savage

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar

David J. Friedman

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

David Friedman

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar

David P. Friedman

Thomas Jefferson University Hospital

View shared research outputs
Top Co-Authors

Avatar

Emmanuel T. Akporiaye

Providence Portland Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge