Network


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

Hotspot


Dive into the research topics where Marka Crittenden is active.

Publication


Featured researches published by Marka Crittenden.


Science Translational Medicine | 2012

Phase 1 study of stereotactic body radiotherapy and interleukin-2--tumor and immunological responses.

Steven K. Seung; Brendan D. Curti; Marka Crittenden; Edwin B. Walker; Todd Coffey; Janet C Siebert; William M. Miller; Roxanne Payne; Lyn A. Glenn; Alexandru Bageac; Walter J. Urba

Stereotactic body radiation therapy enhances tumor response rate to high-dose interleukin-2 in a phase 1 study. Let’s Work Together Despite decades of research into causes and potential therapies for cancer, cancers still account for almost 13% of all deaths every year. It is becoming increasingly clear that monotherapies are not the answer, and combining drugs to improve efficacy and prevent resistance is becoming the norm. However, care must be taken when combining even drugs already in the clinic—two treatments may not necessarily be better than one and may even cause harm. Thus, there is a need for rationally designed combination therapy. Here, Seung et al. conduct a phase 1 trial on one such rationally combined therapy—interleukin-2 (IL-2) and stereotactic body radiation therapy (SBRT). IL-2, an immune activator, has been long used in the clinic either as a single-agent immunotherapy or in combination with various drugs for melanoma and renal cell carcinoma, with limited success. Here, the authors combine high-dose IL-2 with targeted radiation therapy based on clinical observation of enhanced efficacy in patients as well as the still to be proven hypothesis that radiation damage induces tumor antigen release and microenvironment changes that should enhance the immune-activating effects of IL-2. They found that the combination therapy was safe, and, albeit in a small number of patients, appeared to have improved efficacy over IL-2 alone. Intriguingly, they found a greater frequency of proliferating early effector memory T cells in the peripheral blood of these patients. Although studies with more patients and more detailed mechanistic follow-up must be performed, this study suggests that the rational combination of SBRT and IL-2 may improve upon current therapies for metastatic melanoma and renal cell carcinoma. Preclinical models suggest that focal high-dose radiation can make tumors more immunogenic. We performed a pilot study of stereotactic body radiation therapy (SBRT) followed by high-dose interleukin-2 (IL-2) to assess safety and tumor response rate and perform exploratory immune monitoring studies. Patients with metastatic melanoma or renal cell carcinoma (RCC) who had received no previous medical therapy for metastatic disease were eligible. Patients received one, two, or three doses of SBRT (20 Gy per fraction) with the last dose administered 3 days before starting IL-2. IL-2 (600,000 IU per kilogram by means of intravenous bolus infusion) was given every 8 hours for a maximum of 14 doses with a second cycle after a 2-week rest. Patients with regressing disease received up to six IL-2 cycles. Twelve patients were included in the intent-to-treat analysis, and 11 completed treatment per the study design. Response Evaluation Criteria in Solid Tumors criteria were used to assess overall response in nonirradiated target lesions. Eight of 12 patients (66.6%) achieved a complete (CR) or partial response (PR) (1 CR and 7 PR). Six of the patients with PR on computed tomography had a CR by positron emission tomography imaging. Five of seven (71.4%) patients with melanoma had a PR or CR, and three of five (60%) with RCC had a PR. Immune monitoring showed a statistically significantly greater frequency of proliferating CD4+ T cells with an early activated effector memory phenotype (CD3+CD4+Ki67+CD25+FoxP3−CCR7−CD45RA−CD27+CD28+/−) in the peripheral blood of responding patients. SBRT and IL-2 can be administered safely. Because the response rate in patients with melanoma was significantly higher than expected on the basis of historical data, we believe that the combination and investigation of CD4+ effector memory T cells as a predictor of response warrant further study.


Journal of Immunotherapy | 2010

Adjuvant therapy with agonistic antibodies to CD134 (OX40) increases local control after surgical or radiation therapy of cancer in mice.

Michael J. Gough; Marka Crittenden; Maryclare Sarff; Puiyi Pang; Steven K. Seung; John T. Vetto; Hong Ming Hu; William L. Redmond; John M. Holland; Andrew D. Weinberg

The tumor recurrence from residual local or micrometastatic disease remains a problem in cancer therapy. In patients with soft tissue sarcoma and the patients with inoperable nonsmall cell lung cancer, local recurrence is common and significant mortality is caused by the subsequent emergence of metastatic disease. Thus, although the aim of the primary therapy is curative, the outcome may be improved by additional targeting of residual microscopic disease. We display in a murine model that surgical removal of a large primary sarcoma results in local recurrence in approximately 50% of animals. Depletion of CD8 T cells results in local recurrence in 100% of animals, indicating that these cells are involved in the control of residual disease. We further show that systemic adjuvant administration of αOX40 at surgery eliminates local recurrences. In this model, αOX40 acts to directly enhance tumor antigen-specific CD8 T-cell proliferation in the lymph node draining the surgical site, and results in increased tumor antigen-specific cytotoxicity in vivo. These results are also corroborated in a murine model of hypofractionated radiation therapy of lung cancer. Administration of αOX40 in combination with radiation significantly extended the survival compared with either agent alone, and resulted in a significant proportion of long-term tumor-free survivors. We conclude that αOX40 increases tumor antigen-specific CD8 T-cell cytotoxic activity resulting in improved endogenous immune control of residual microscopic disease, and we propose that adjuvant αOX40 administration may be a valuable addition to surgical and radiation therapy for cancer.


Seminars in Oncology | 2010

Signaling Through OX40 Enhances Antitumor Immunity

Shawn M. Jensen; Levi D. Maston; Michael J. Gough; Carl E. Ruby; William L. Redmond; Marka Crittenden; Yuhuan Li; Sachin Puri; Christian H. Poehlein; Nicholas P. Morris; Magdalena Kovacsovics-Bankowski; Tarsem Moudgil; Chris Twitty; Edwin B. Walker; Hong-Ming Hu; Walter J. Urba; Andrew D. Weinberg; Brendan D. Curti; Bernard A. Fox

The existence of tumor-specific T cells, as well as their ability to be primed in cancer patients, confirms that the immune response can be deployed to combat cancer. However, there are obstacles that must be overcome to convert the ineffective immune response commonly found in the tumor environment to one that leads to sustained destruction of tumor. Members of the tumor necrosis factor (TNF) superfamily direct diverse immune functions. OX40 and its ligand, OX40L, are key TNF members that augment T-cell expansion, cytokine production, and survival. OX40 signaling also controls regulatory T-cell differentiation and suppressive function. Studies over the past decade have demonstrated that OX40 agonists enhance antitumor immunity in preclinical models using immunogenic tumors; however, treatment of poorly immunogenic tumors has been less successful. Combining strategies that prime tumor-specific T cells together with OX40 signaling could generate and maintain a therapeutic antitumor immune response.


Human Gene Therapy | 2002

Cells as vehicles for cancer gene therapy: the missing link between targeted vectors and systemic delivery?

Kevin J. Harrington; Luis Álvarez-Vallina; Marka Crittenden; Michael J. Gough; Heung Chong; Rosa Maria Diaz; Georges Vassaux; Nicholas R. Lemoine; Richard G. Vile

Systemic administration of currently manufactured viral stocks has not so far achieved sufficient circulating titers to allow therapeutic targeting of metastatic disease. This is due to low initial viral titers, immune inactivation, nonspecific adhesion, and loss of particles. One way to exploit the elegant molecular manipulations that have been made to increase vector targeting is to protect these vectors until they reach the local sites of tumor growth. Various cell types home preferentially to tumors and can be loaded with the constructs required to produce targeted vectors. Here we discuss the potential of using such cell carriers to chaperone precious vectors directly to the tumors. The vectors can incorporate mechanisms to achieve tumor site-inducible expression, along with tumor cell-specific expression of the therapeutic gene and/or replicating viral genomes that would be released at the tumor. In this way, the great advances that have so far been made with the engineering of vector tropisms might be genuinely exploited and converted into clinical benefit.


Seminars in Radiation Oncology | 2015

Current Clinical Trials Testing Combinations of Immunotherapy and Radiation

Marka Crittenden; Holbrook Kohrt; Ronald Levy; Jennifer Jones; Kevin Camphausen; Adam P. Dicker; Sandra Demaria; Silvia C. Formenti

Preclinical evidence of successful combinations of ionizing radiation with immunotherapy has inspired testing the translation of these results to the clinic. Interestingly, the preclinical work has consistently predicted the responses encountered in clinical trials. The first example came from a proof-of-principle trial started in 2001 that tested the concept that growth factors acting on antigen-presenting cells improve presentation of tumor antigens released by radiation and induce an abscopal effect. Granulocyte-macrophage colony-stimulating factor was administered during radiotherapy to a metastatic site in patients with metastatic solid tumors to translate evidence obtained in a murine model of syngeneic mammary carcinoma treated with cytokine FLT-3L and radiation. Subsequent clinical availability of vaccines and immune checkpoint inhibitors has triggered a wave of enthusiasm for testing them in combination with radiotherapy. Examples of ongoing clinical trials are described in this report. Importantly, most of these trials include careful immune monitoring of the patients enrolled and will generate important data about the proimmunogenic effects of radiation in combination with a variety of immune modulators, in different disease settings. Results of these studies are building a platform of evidence for radiotherapy as an adjuvant to immunotherapy and encourage the growth of this novel field of radiation oncology.


Immunology | 2005

Intratumoral immunotherapy: Using the tumour against itself

Marka Crittenden; Uma Thanarajasingam; Richard Vile; Michael J. Gough

Diverse immunotherapy approaches have achieved success in controlling individual aspects of immune responses in animal models. Transfer of such immunotherapies to clinical trials has obtained some success in patients, with clinical responses observed or effective antigen specific immune responses achieved, but has had limited impact on patient survival. Key elements required to generate de novo cell‐mediated antitumour immune responses in vivo include recruitment of antigen‐presenting cells to the tumour site, loading these cells with antigen, and their migration and maturation to full antigen‐presenting function. In addition, it is essential for antigen‐specific T cells to locate the tumour to mediate cytotoxicity, emphasizing the need for local inflammation to target effector cell recruitment. We review those therapies that involve the tumour site as a target and source of antigen for the initiation of immune responses, and discuss strategies to generate and co‐ordinate an optimal cell‐mediated immune response to control tumours locally.


Cancer Research | 2016

Radiotherapy Combined with Novel STING-Targeting Oligonucleotides Results in Regression of Established Tumors

Jason R. Baird; David Friedman; Benjamin Cottam; Thomas W. Dubensky; David B. Kanne; Shelly Bambina; Keith S. Bahjat; Marka Crittenden; Michael J. Gough

Cytotoxic therapies prime adaptive immune responses to cancer by stimulating the release of tumor-associated antigens. However, the tumor microenvironment into which these antigens are released is typically immunosuppressed, blunting the ability to initiate immune responses. Recently, activation of the DNA sensor molecule STING by cyclic dinucleotides was shown to stimulate infection-related inflammatory pathways in tumors. In this study, we report that the inflammatory pathways activated by STING ligands generate a powerful adjuvant activity for enhancing adaptive immune responses to tumor antigens released by radiotherapy. In a murine model of pancreatic cancer, we showed that combining CT-guided radiotherapy with a novel ligand of murine and human STING could synergize to control local and distant tumors. Mechanistic investigations revealed T-cell-independent and TNFα-dependent hemorrhagic necrosis at early times, followed by later CD8 T-cell-dependent control of residual disease. Clinically, STING was found to be expressed extensively in human pancreatic tumor and stromal cells. Our findings suggest that this novel STING ligand could offer a potent adjuvant for leveraging radiotherapeutic management of pancreatic cancer.


Journal of Immunology | 2005

Gene Therapy to Manipulate Effector T Cell Trafficking to Tumors for Immunotherapy

Michael J. Gough; Marka Crittenden; Uma Thanarajasingam; Luis Sanchez-Perez; Jill Thompson; Dragan Jevremovic; Richard G. Vile

Strategies that generate tumor Ag-specific effector cells do not necessarily cure established tumors. We hypothesized that the relative efficiency with which tumor-specific effector cells reach the tumor is critical for therapy. We demonstrate in this study that activated T cells respond to the chemokine CCL3, both in vitro and in vivo, and we further demonstrate that expression of CCL3 within tumors increases the effector T cell infiltrate in those tumors. Importantly, we show that adenoviral gene transfer to cause expression of CCL3 within B16ova tumors in vivo increases the efficacy of adoptive transfer of tumor-specific effector OT1 T cells. We additionally demonstrate that such therapies result in endogenous immune responses to tumor Ags that are capable of protecting animals against subsequent tumor challenge. Strategies that modify the “visibility” of tumors have the potential to significantly enhance the efficacy of both vaccine and adoptive transfer therapies currently in development.


Journal of Immunology | 2004

MIP-3α Transfection into a Rodent Tumor Cell Line Increases Intratumoral Dendritic Cell Infiltration but Enhances (Facilitates) Tumor Growth and Decreases Immunogenicity

Bernard Bonnotte; Marka Crittenden; Nicolas Larmonier; Michael J. Gough; Richard Vile

Dendritic cells are powerful APCs for activation of specific antitumor T lymphocytes. To present tumor Ags efficiently, they have first to migrate to the tumor site, engulf Ag, and then process them. To attract immature DCs to the tumor site, we transfected tumor cells with MIP-3α which is strongly chemotactic for DCs. Surprisingly, MIP-3α-transfected tumor cells grew faster than the mock-transfected tumor cells. Histological analysis and tumor dissociation confirmed that the MIP-3α-transfected tumors contain three to four times more DCs than mock-transfected tumors. FACS analysis of the intratumor DCs showed that they were predominantly immature. Functional analysis showed that the alloreactivity mediated by these infiltrating MIP-3α-transfected tumor DCs is strongly reduced. In conclusion, MIP-3α is an efficient chemokine for attracting DCs in vivo, but the high density of DCs in the tumor site injection is not a sufficient condition to induce an immune response. Furthermore, this attraction of immature DCs may always have an adverse effect by inducing a tolerance to the tumor cells.


PLOS ONE | 2012

Expression of NF-κB p50 in Tumor Stroma Limits the Control of Tumors by Radiation Therapy

Marka Crittenden; Benjamin Cottam; Talicia Savage; Cynthia Nguyen; Pippa Newell; Michael J. Gough

Radiation therapy aims to kill cancer cells with a minimum of normal tissue toxicity. Dying cancer cells have been proposed to be a source of tumor antigens and may release endogenous immune adjuvants into the tumor environment. For these reasons, radiation therapy may be an effective modality to initiate new anti-tumor adaptive immune responses that can target residual disease and distant metastases. However, tumors engender an environment dominated by M2 differentiated tumor macrophages that support tumor invasion, metastases and escape from immune control. In this study, we demonstrate that following radiation therapy of tumors in mice, there is an influx of tumor macrophages that ultimately polarize towards immune suppression. We demonstrate using in vitro models that this polarization is mediated by transcriptional regulation by NFκB p50, and that in mice lacking NFκB p50, radiation therapy is more effective. We propose that despite the opportunity for increased antigen-specific adaptive immune responses, the intrinsic processes of repair following radiation therapy may limit the ability to control residual disease.

Collaboration


Dive into the Marka Crittenden's collaboration.

Top Co-Authors

Avatar

Michael J. Gough

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
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

Brendan D. Curti

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar

Keith S. Bahjat

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

R. Bryan Bell

Providence Portland Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge