Network


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

Hotspot


Dive into the research topics where Melissa Walker is active.

Publication


Featured researches published by Melissa Walker.


Clinical Cancer Research | 2010

Phase I and Pharmacokinetic Studies of CYT-6091, a Novel PEGylated Colloidal Gold-rhTNF Nanomedicine

Steven K. Libutti; Giulio F. Paciotti; Adriana A. Byrnes; H. Richard Alexander; William E. Gannon; Melissa Walker; Geoffrey Seidel; Nargiza Yuldasheva; Lawrence Tamarkin

Purpose: A novel nanomedicine, CYT-6091, constructed by simultaneously binding recombinant human tumor necrosis factor alpha (rhTNF) and thiolyated polyethylene glycol to the surface of 27-nm colloidal gold particles, was tested in a phase I dose escalation clinical trial in advanced stage cancer patients. Experimental Design: CYT-6091, whose dosing was based on the amount of rhTNF in the nanomedicine, was injected intravenously, and 1 cycle of treatment consisted of 2 treatments administered 14 days apart. Results: Doses from 50 μg/m2 to 600 μg/m2 were well tolerated, and no maximum tolerated dose (MTD) was reached, as the highest dose exceeded the target dosage of 1-mg rhTNF per treatment, exceeding the previous MTD for native rhTNF by 3-fold. The first 2 patients on the study, each receiving 50 μg/m2, did not receive any prophylactic antipyretics or H2 blockade. A predicted, yet controllable fever occurred in these patients, so all subsequently treated patients received prophylactic antipyretics and H2 blockers. However, even at the highest dose rhTNFs dose-limiting toxic effect of hypotension was not seen. Using electron microscopy to visualize nanoparticles of gold in patient biopsies of tumor and healthy tissue showed that patient biopsies taken 24 hours after treatment had nanoparticles of gold in tumor tissue. Conclusions: These data indicate that rhTNF formulated as CYT-6091 may be administered systemically at doses of rhTNF that were previously shown to be toxic and that CYT-6091 may target to tumors. Future clinical studies will focus on combining CYT-6091 with approved chemotherapies for the systemic treatment of nonresectable cancers. Clin Cancer Res; 16(24); 6139–49. ©2010 AACR.


Journal of Surgical Oncology | 2014

Impact of Maximal Cytoreductive Surgery Plus Regional Heated Intraperitoneal Chemotherapy (HIPEC) on Outcome of Patients With Peritoneal Carcinomatosis of Gastric Origin: Results of the GYMSSA Trial

Udo Rudloff; Russell C. Langan; John E. Mullinax; Joal D. Beane; Seth M. Steinberg; Tatiana Beresnev; Carole C. Webb; Melissa Walker; Mary Ann Toomey; David S. Schrump; Prakash Pandalai; Alexander Stojadinovic; Itzhak Avital

A prospective randomized trial was conducted to compare the impact of systemic chemotherapy versus multi‐modality therapy (complete cytoreductive surgery (CRS), hyperthermic intraperitoneal chemotherapy (HIPEC), and systemic chemotherapy) on overall survival (OS) in patients with gastric carcinomatosis.


Trials | 2009

The GYMSSA trial: a prospective randomized trial comparing gastrectomy, metastasectomy plus systemic therapy versus systemic therapy alone

Sid P. Kerkar; Clinton D. Kemp; Austin Duffy; Udai S. Kammula; David S. Schrump; King F. Kwong; Martha Quezado; Barry R. Goldspiel; Aradhana M. Venkatesan; Ann Berger; Melissa Walker; Mary Ann Toomey; Seth M. Steinberg; Guiseppe Giaccone; Steven A. Rosenberg; Itzhak Avital

BackgroundThe standard of care for metastatic gastric cancer (MGC) is systemic chemotherapy which leads to a median survival of 6-15 months. Survival beyond 3 years is rare. For selected groups of patients with limited MGC, retrospective studies have shown improved overall survival following gastrectomy and metastasectomies including peritoneal stripping with continuous hyperthermic peritoneal perfusion (CHPP), liver resection, and pulmonary resection. Median survival after liver resection for MGC is up to 34 months, with a five year survival rate of 24.5%. Similarly, reported median survival after pulmonary resection of MGC is 21 months with long term survival of greater than 5 years a possibility. Several case reports and small studies have documented evidence of long-term survival in select individuals who undergo CHPP for MGC.DesignThe GYMSSA trial is a prospective randomized trial for patients with MGC. It is designed to compare two therapeutic approaches: gastrectomy with metastasectomy plus systemic chemotherapy (GYMS) versus systemic chemotherapy alone (SA). Systemic therapy will be composed of the FOLFOXIRI regimen. The aim of the study is to evaluate overall survival and potential selection criteria to determine those patients who may benefit from surgery plus systemic therapy. The study will be conducted by the Surgery Branch at the National Cancer Institute (NCI), National Institutes of Health (NIH) in Bethesda, Maryland. Surgeries and followup will be done at the NCI, and chemotherapy will be given by either the local oncologist or the medical oncology branch at NCI.Trial RegistrationClinicalTrials.gov ID. NCT00941655


United European gastroenterology journal | 2015

A phase II study of TRC105 in patients with hepatocellular carcinoma who have progressed on sorafenib

Austin Duffy; Susanna Varkey Ulahannan; Liang Cao; Osama E. Rahma; Oxana V. Makarova-Rusher; David E. Kleiner; Suzanne Fioravanti; Melissa Walker; S Carey; Yunkai Yu; Aradhana M. Venkatesan; Baris Turkbey; Peter L. Choyke; Jane B. Trepel; Kc Bollen; Seth M. Steinberg; William D. Figg; Tim F. Greten

Background Endoglin is an endothelial cell membrane receptor essential for angiogenesis and highly expressed on the vasculature of many tumor types, including hepatocellular carcinoma (HCC). TRC105 is a chimeric IgG1 anti-CD105 monoclonal antibody that inhibits angiogenesis and tumor growth by endothelial cell growth inhibition, ADCC and apoptosis, and complements VEGF inhibitors. Objective The aim of this phase II study was to evaluate the efficacy of anti-endoglin therapy with TRC105 in patients with advanced HCC, post-sorafenib. Methods Patients with HCC and compensated liver function (Childs-Pugh A/B7), ECOG 0/1, were enrolled to a single-arm, phase II study of TRC105 15 mg/kg IV every two weeks. Patients must have progressed on or been intolerant of prior sorafenib. A Simon optimal two-stage design was employed with a 50% four-month PFS target for progression to the second stage. Correlative biomarkers evaluated included DCE-MRI as well as plasma levels of angiogenic biomarkers and soluble CD105. Results A total accrual of 27 patients was planned. However, because of lack of efficacy and in accordance with the Simon two-stage design, 11 patients were enrolled. There were no grade 3/4 treatment-related toxicities. Most frequent toxicities were headache (G2; N = 3) and epistaxis (G1; N = 4). One patient had a confirmed partial response by standard RECIST criteria and biologic response on DCE-MRI but the four-month PFS was insufficient to proceed to the second stage of the study. Conclusions: TRC105 was well tolerated in this HCC population following sorafenib. Although there was evidence of clinical activity, this did not meet prespecified criteria to proceed to the second stage. TRC105 development in HCC continues as combination therapy with sorafenib.


Trials | 2011

Regional Chemotherapy in Locally Advanced Pancreatic Cancer: RECLAP Trial

Jeremy L. Davis; Prakash Pandalai; R. Taylor Ripley; Russell C. Langan; Seth M. Steinberg; Melissa Walker; Mary Ann Toomey; E. Levy; Itzhak Avital

BackgroundPancreatic cancer is the fourth leading cause of cancer death in the United States. Surgery offers the only chance for cure. However, less than twenty percent of patients are considered operative candidates at the time of diagnosis. A common reason for being classified as unresectable is advanced loco-regional disease.A review of the literature indicates that almost nine hundred patients with pancreatic cancer have received regional chemotherapy in the last 15 years. Phase I studies have shown regional administration of chemotherapy to be safe. The average reported response rate was approximately 26%. The average 1-year survival was 39%, with an average median survival of 9 months. Of the patients that experienced a radiographic response to therapy, 78 (78/277, 28%) patients underwent exploratory surgery following regional chemotherapy administration; thirty-two (41%) of those patients were amenable to pancreatectomy. None of the studies performed analyses to identify factors predicting response to regional chemotherapy.Progressive surgical techniques combined with current neoadjuvant chemoradiotherapy strategies have already yielded emerging support for a multimodality approach to treatment of advanced pancreatic cancer.Intravenous gemcitabine is the current standard treatment of pancreatic cancer. However, >90% of the drug is secreted unchanged affecting toxicity but not the cancer per se. Gemcitabine is converted inside the cell into its active drug form in a rate limiting reaction. We hypothesize that neoadjuvant regional chemotherapy with continuous infusion of gemcitabine will be well tolerated and may improve resectability rates in cases of locally advanced pancreatic cancer.DesignThis is a phase I study designed to evaluate the feasibility and toxicity of super-selective intra-arterial administration of gemcitabine in patients with locally advanced, unresectable pancreatic adenocarcinoma. Patients considered unresectable due to locally advanced pancreatic cancer will receive super-selective arterial infusion of gemcitabine over 24 hours via subcutaneous indwelling port. Three to six patients will be enrolled per dose cohort, with seven cohorts, plus an additional six patients at the maximum tolerated dose; accrual is expected to last 36 months. Secondary objectives will include the determination of progression free and overall survival, as well as the conversion rate from unresectable to potentially resectable pancreatic cancer.Trial RegistrationClinicalTrials.gov ID: NCT01294358


International Journal of Cancer | 2016

Modulation of tumor eIF4E by antisense inhibition: A phase I/II translational clinical trial of ISIS 183750 – an antisense oligonucleotide against eIF4E – in combination with irinotecan in solid tumors and irinotecan‐refractory colorectal cancer

Austin Duffy; Oxana V. Makarova-Rusher; Susanna Varkey Ulahannan; Osama E. Rahma; Suzanne Fioravanti; Melissa Walker; S. Abdullah; Mark Raffeld; Victoria L. Anderson; Nadine Abi-Jaoudeh; E. Levy; Bradford J. Wood; Su Jae Lee; Yasushi Tomita; Jane B. Trepel; Seth M. Steinberg; A.S. Revenko; A.R. MacLeod; Cody J. Peer; William D. Figg; Tim F. Greten

The eukaryotic translation initiation factor 4E (eIF4E) is a potent oncogene that is found to be dysregulated in 30% of human cancer, including colorectal carcinogenesis (CRC). ISIS 183750 is a second‐generation antisense oligonucleotide (ASO) designed to inhibit the production of the eIF4E protein. In preclinical studies we found that EIF4e ASOs reduced expression of EIF4e mRNA and inhibited proliferation of colorectal carcinoma cells. An additive antiproliferative effect was observed in combination with irinotecan. We then performed a clinical trial evaluating this combination in patients with refractory cancer. No dose‐limiting toxicities were seen but based on pharmacokinetic data and tolerability the dose of irinotecan was reduced to 160 mg/m2 biweekly. Efficacy was evaluated in 15 patients with irinotecan‐refractory colorectal cancer. The median time of disease control was 22.1 weeks. After ISIS 183750 treatment, peripheral blood levels of eIF4E mRNA were decreased in 13 of 19 patients. Matched pre‐ and posttreatment tumor biopsies showed decreased eIF4E mRNA levels in five of nine patients. In tumor tissue, the intracellular and stromal presence of ISIS 183750 was detected by IHC in all biopsied patients. Although there were no objective responses stable disease was seen in seven of 15 (47%) patients who were progressing before study entry, six of whom were stable at the time of the week 16 CT scan. We were also able to confirm through mandatory pre‐ and posttherapy tumor biopsies penetration of the ASO into the site of metastasis.


Journal for ImmunoTherapy of Cancer | 2014

Paired tumor biopsy analysis and safety data from a pilot study evaluating Tremelimumab - a monoclonal antibody against CTLA-4 - in combination with ablative therapy in patients with hepatocellular carcinoma (HCC)

Austin Duffy; Sid P. Kerkar; David E. Kleiner; Susanna Varkey Ulahannan; Metin Kurtoglu; Oxana Rusher; Suzanne Fioravanti; Melissa Walker; William D. Figg; Kathryn Compton; Aradhana M. Venkatesan; Nadine Abi-Jaoudeh; Brad J. Wood; Tim F. Greten

Meeting abstracts Tremelimumab is a fully human monoclonal antibody that binds to CTLA-4 expressed on the surface of activated T lymphocytes and results in inhibition of B7-CTLA-4-mediated down regulation of T cell activation. Both transcatheter arterial chemoembolization (TACE) and radiofrequency


Journal of Clinical Oncology | 2009

Results of a completed phase I clinical trial of CYT-6091: A pegylated colloidal gold-TNF nanomedicine

Steven K. Libutti; Giulio F. Paciotti; L. Myer; R. Haynes; W. Gannon; Melissa Walker; G. Seidel; A. Byrnes; N. Yuldasheva; Lawrence Tamarkin


Journal of Clinical Oncology | 2016

Tremelimumab, a monoclonal antibody against CTLA-4, in combination with subtotal ablation (trans-catheter arterial chemoembolization [TACE], radiofrequency ablation [RFA] or cryoablation) in patients with hepatocellular carcinoma (HCC) and biliary tract carcinoma (BTC).

Austin Duffy; Oxana V. Makarova-Rusher; Drew Pratt; David E. Kleiner; Suzanne Fioravanti; Melissa Walker; Stephanie Carey; William D. Figg; Seth M. Steinberg; Victoria L. Anderson; E. Levy; Venkatesh Krishnasamy; Bradford J. Wood; Tim F. Greten


Journal of Clinical Oncology | 2016

A pilot study of AMP-224, a PD-L2 Fc fusion protein, in combination with stereotactic body radiation therapy (SBRT) in patients with metastatic colorectal cancer.

Austin G. Duffy; Oxana V. Makarova-Rusher; Drew Pratt; David E. Kleiner; Suzanne Fioravanti; Melissa Walker; Stephanie Carey; William D. Figg; Seth M. Steinberg; Victoria L. Anderson; Elliot Levy; Venkatesh Krishnasamy; Bradford J. Wood; J Jones; Deborah Citrin; Tim F. Greten

Collaboration


Dive into the Melissa Walker's collaboration.

Top Co-Authors

Avatar

Suzanne Fioravanti

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tim F. Greten

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Seth M. Steinberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

William D. Figg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Austin G. Duffy

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

David E. Kleiner

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Bradford J. Wood

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Aradhana M. Venkatesan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Austin Duffy

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge