Network


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

Hotspot


Dive into the research topics where Kari Kendra is active.

Publication


Featured researches published by Kari Kendra.


Journal of Clinical Oncology | 2004

Phase I Clinical Trial of the Immunocytokine EMD 273063 in Melanoma Patients

David M. King; Mark R. Albertini; Heidi Schalch; Jacquelyn A. Hank; Jacek Gan; Jean E. Surfus; David M. Mahvi; Joan H. Schiller; Thomas F. Warner; Kyung Mann Kim; Jens C. Eickhoff; Kari Kendra; Ralph A. Reisfeld; Stephen D. Gillies; Paul M. Sondel

PURPOSE To evaluate the safety, toxicity, in vivo immunologic activation, and maximum-tolerated dose (MTD) of EMD 273063 (hu14.18-IL-2) in patients with metastatic melanoma. PATIENTS AND METHODS Thirty-three patients were treated with EMD 273063, a humanized anti-GD2 monoclonal antibody (mAb) linked to interleukin-2 (IL-2). EMD 273063 was given as a 4-hour intravenous infusion on days 1, 2, and 3 of week 1. Patients with stabilization or regression of disease could receive a second course of treatment at week 5. Dose levels evaluated were 0.8, 1.6, 3.2, 4.8, 6.0, and 7.5 mg/m2/d. RESULTS Nineteen of 33 patients completed course 1 with stable disease and went on to receive course 2. Eight patients had stable disease on completion of course 2. Grade 3 adverse events included hypophosphatemia (11 patients), hyperglycemia (three patients), hypotension (two patients), thrombocytopenia (one patient), hypoxia (three patients), elevated hepatic transaminases (two patients), and hyperbilirubinemia (one patient). Opioids were required for treatment-associated arthralgias and/or myalgias during 17 of 52 treatment courses. No grade 4 adverse events were observed. Dose-limiting toxicities at the MTD included hypoxia, hypotension, and elevations in AST/ALT. Grade 3 toxicities were anticipated based on prior studies of IL-2 or anti-GD2 mAbs, and all resolved. Immune activation was induced, as measured by lymphocytosis, increased peripheral-blood natural killer activity, and cell numbers, and increased serum levels of the soluble alpha chain of the IL-2 receptor complex. CONCLUSION Treatment with the immunocytokine EMD 273063 induced immune activation and was associated with reversible clinical toxicities at the MTD of 7.5 mg/m2/d in melanoma patients.


Cancer Immunology, Immunotherapy | 1999

Pharmacokinetics and stability of the ch14.18-interleukin-2 fusion protein in mice.

Kari Kendra; Jacek Gan; Melody Ricci; Jean E. Surfus; Anisa Shaker; Michael Super; Jami D. Frost; Alexander L. Rakhmilevich; Jacquelyn A. Hank; Stephen D. Gillies; Paul M. Sondel

Abstract The fusion protein formed from ch14.18 and interleukin-2 (ch14.18–IL-2), shown to exhibit antitumor efficacy in mouse models, consists of IL-2 genetically linked to each heavy chain of the ch14.18 chimeric anti-GD2 monoclonal antibody. The purpose of this study was to determine the pharmacokinetics of ch14.18–IL-2 in mice and assess its stability in murine serum. Following i.v. injection, the fusion protein was found to have a terminal half-life of 4.1 h. Detection of IL-2 following injection of the ch14.18–IL-2 fusion protein showed a similar half-life, indicating that the fusion protein prolongs the circulatory half-life of IL-2. Detection of human IgG1 following injection of ch14.18–IL-2 showed a terminal half-life of 26.9 h. These data suggested that the native fusion protein is being altered in vivo, resulting in a somewhat rapid loss of detectable IL-2, despite prolonged circulation of its immunoglobulin components. In vitro incubation of the ch14.18–IL-2 fusion protein in pooled mouse serum at 37 °C for 48 h resulted in a loss of its IL-2 component, as detected in enzyme-linked immunosorbent assay systems and in proliferation assays. Polyacrylamide gel electrophoresis and Western blot analysis of the fusion protein incubated in mouse serum at 37 °C indicated that the ch14.18–IL-2 is cleaved, resulting in a loss of the 67-kDa band (representing the IL-2 linked to the IgG1 heavy chain) and the detection of a band of more than 50 kDa, slightly heavier than the IgG1 heavy chain itself. This suggests that the fusion protein is being cleaved in vitro within the IL-2 portion of the molecule. These studies show that (1) ch14.18–IL-2 prolongs the circulatory half-life of IL-2 (compared to that of soluble IL-2) and (2) the in vivo clearance of the fusion protein occurs more rapidly than the clearance of the ch14.18 antibody itself, possibly reflecting in vivo cleavage within the IL-2 portion of the molecule, resulting in loss of IL-2 activity.


Journal of Immunotherapy | 2011

A phase 2 trial of bevacizumab and high-dose interferon alpha 2B in metastatic melanoma.

Valerie P. Grignol; Thomas Olencki; Kiran V. Relekar; Cynthia Taylor; Amanda Kibler; Cheryl Kefauver; Lai Wei; Michael J. Walker; Helen X. Chen; Kari Kendra; William E. Carson

Bevacizumab is a humanized recombinant monoclonal antibody that neutralizes vascular endothelial growth factor, an agent with proangiogenic effects in melanoma. Interferon alpha (IFN-&agr;) has antiangiogenic properties through its ability to downregulate basic-fibroblast growth factor levels. We hypothesized that the coadministration of these agents would lead to tumor regression. Patients with metastatic melanoma received bevacizumab 15 mg/kg intravenously on day 1 of the 2-week cycle. IFN-&agr; was administered thrice weekly at 5 MU/m2 subcutaneously during cycle 1 and was increased to 10 MU/m2 during cycle 2. Patients were restaged every 6 cycles. Patients with stable disease or a response continued with therapy. Baseline serum vascular endothelial growth factor and fibroblast growth factor were measured. Twenty-five patients were accrued. Mean age was 58.4 years. Eleven patients required IFN-&agr; dose reductions due to toxicity. Common grade 3 toxicities associated with IFN-&agr; included fatigue and myalgia. Bevacizumab administration was associated with grade 2-3 proteinuria in 6 patients. Grade 4 adverse events were pulmonary embolus (1), myocardial infarction (1), and stroke (1). Six patients had a partial response, and 5 patients exhibited stable disease that lasted more than 24 weeks (range: 30 to 122 wk). Median progression-free survival and overall survival were 4.8 and 17 months, respectively. Significantly lower fibroblast growth factor levels were observed in patients with a partial response compared to those with stable or progressive disease (P=0.040). Administration of bevacizumab with IFN led to a clinical response in 24% of patients with stage IV melanoma and stabilization of disease in another 20% of patients. This regimen has activity in advanced melanoma.


Journal of Clinical Oncology | 2008

Randomized Phase II Adjuvant Trial of Dose-Dense Docetaxel Before or After Doxorubicin Plus Cyclophosphamide in Axillary Node-Positive Breast Cancer

Shannon Puhalla; Ewa Mrozek; Donn C. Young; Susan Ottman; Anne McVey; Kari Kendra; Nancy Merriman; Mark Knapp; Taral Patel; Mark E. Thompson; James Maher; Timothy David Moore; Charles L. Shapiro

PURPOSE An anthracycline-based combination followed by, or combined with, a taxane is the sequence used in most adjuvant chemotherapy regimens. We hypothesized that administering the taxane before the anthracycline combination would be associated with fewer dose reductions and delays than the reverse sequence. To test this hypothesis, a randomized phase II multicenter adjuvant chemotherapy trial was performed. PATIENTS AND METHODS Fifty-six patients with axillary node-positive, nonmetastatic breast cancer were randomly assigned either to group A (docetaxel [DOC] 75 mg/m(2) intravenously [IV] every 14 days for four cycles followed by doxorubicin 60 mg/m(2) and cyclophosphamide 600 mg/m(2) [AC] IV every 14 days for four cycles); or to group B (AC followed by DOC) at the identical doses and schedule. Pegfilgrastim 6 mg subcutaneous injection was administered 1 day after the chemotherapy in all treatment cycles. The primary objective was to administer DOC without dose reductions or delays before or after AC and calculate the relative dose intensity (RDI) of DOC and AC. RESULTS The majority of toxicities were grade 0 to 2 irrespective of sequence. The RDI for DOC was 0.96 and 0.82, respectively, in groups A (DOC followed by AC) and B (AC followed by DOC), with more frequent dose reductions occurring in group B (46% v 18%). The RDI for AC was 0.95 and 0.98 in groups A and B, respectively. CONCLUSION The administration of DOC before AC results in fewer DOC dose reductions and a higher RDI than the reverse sequence. Larger trials evaluating the sequence of DOC before anthracyclines are justified.


American Journal of Clinical Oncology | 2011

A pilot study of bevacizumab and interferon-α2b in ocular melanoma.

Kristan D. Guenterberg; Valerie P. Grignol; Kiran V. Relekar; Kimberly A. Varker; Helen X. Chen; Kari Kendra; Thomas Olencki; William E. Carson

Objectives: We hypothesized that administration of bevacizumab, a monoclonal antibody that neutralizes vascular endothelial growth factor, in combination with high-dose interferon-alpha2b (IFN-&agr;2b), an inhibitor of basic fibroblast growth factor, would have clinical activity in patients with metastatic ocular melanoma. Methods: Patients with metastatic ocular melanoma received bevacizumab (15 mg/kg intravenously every 2 weeks) plus IFN-&agr;2b (5 MU/m2 subcutaneously 3 times weekly for 2 weeks followed by a dose of 10 MU/m2 subcutaneously thereafter). Patients exhibiting a clinical response or stabilization of disease were treated until disease progression. Results: In this pilot study, 5 patients were treated (3 men, 2 women) with a mean age of 63.8 years (range, 53–71 years). Overall, the regimen was well-tolerated. The following adverse events were noted: grade 3 dyspnea (2 patients), grade 3 and 4 fatigue (2), grade 3 muscle weakness (1), grade 3 anorexia (1), grade 1 and 2 proteinuria (2), and grade 3 diarrhea (1). All adverse events resolved with a treatment holiday or dose reduction. One patient had reduction in tumor burden of 23% by Response Evaluation Criteria in Solid Tumors criteria and 2 patients had stabilization of disease lasting 28 and 36 weeks, respectively. Two patients failed to respond and progressed after 6 and 7 weeks of therapy. Conclusion: Bevacizumab and IFN-&agr;2b were well tolerated in this patient population, and clinical activity was observed. Further study of high-dose IFN-&agr;2b in combination with bevacizumab in this setting is warranted.


Journal of Immunotherapy | 2014

A phase I trial of bortezomib and interferon-α-2b in metastatic melanoma.

Joseph Markowitz; Eric Luedke; Valerie P. Grignol; Erinn M. Hade; Bonnie Paul; Bethany L. Mundy-Bosse; Taylor R. Brooks; Thao-Vi Dao; Sri Vidya Kondalasula; Gregory B. Lesinski; Thomas Olencki; Kari Kendra; William E. Carson

The possibility that cytokine administration could enhance the antitumor effects of proteasome inhibition was explored. It was found that coadministration of bortezomib and interferon-&agr; (IFN-&agr;) induced synergistic apoptosis in human melanoma cell lines and prolonged survival in a murine model of melanoma. A phase I study was conducted to determine the tolerability and the maximum tolerated dose of bortezomib when administered in combination with IFN-&agr;-2b to patients with metastatic melanoma. Patients were treated on a 5-week cycle. In week 1 of cycle 1, patients received 5 million U/m2 IFN-&agr; subcutaneously thrice weekly. During weeks 2–4 of cycle 1, bortezomib was administered intravenously weekly along with IFN-&agr; thrice weekly. There was a treatment break during week 5. After cycle 1, bortezomib was administered in combination with IFN-&agr;. Bortezomib was administered in escalating doses (1.0, 1.3, or 1.6 mg/m2) to cohorts of 3 patients. Sixteen patients were treated (8 women, 8 men; median age 59 y). Common grade 3 toxicities included fatigue (5), vomiting (3), and diarrhea (3). Grade 4 toxicities included fatigue (3) and lymphopenia (1). The maximum tolerated dose for bortezomib was 1.3 mg/m2. One patient had a partial response, and 7 had stable disease. Progression-free survival was 2.5 months, and overall survival was 10.3 months. Bortezomib administration did not augment the ability of IFN-&agr; to induce phosphorylation of STAT1 in circulating immune cells; however, it did lead to reduced plasma levels of proangiogenic cytokines. The combination of bortezomib and IFN-&agr; can be safely administered to melanoma patients.


Journal of Immunotherapy | 1999

In vivo binding and antitumor activity of Ch14.18.

Kari Kendra; Vera Malkovska; Mark Allen; Jose Guzman; Mark R. Albertini

The melanoma reactive chimeric 14.18 (ch14.18) antibody can mediate enhanced in vitro lysis of human M-21 melanoma cells. This study analyzes the antitumor effects and the in vivo binding of ch14.18 antibody with M-21 melanoma cells in severe combined immunodeficiency (SCID) mice. Outgrowth of tumors was prevented in 6/6 animals by the simultaneous subcutaneous injection of peripheral blood mononuclear cells (PBMC) [3 x 10(6) cells (2 animals); 10 x 10(6) cells (2 animals); and 30 x 10(6) cells (2 animals)], with 0.5 mg ch14.18, 1,500 U interleukin 2 (IL-2), and 10(6) M-21 cells. In contrast, 7 of 7 control mice that received M-21 cells alone, 7 of 7 mice that received M-21 cells and ch14.18, and 5 of 6 mice that received M-21 cells plus PBMC plus IL-2, grew subcutaneous tumors. The in vivo localization of ch14.18 was then evaluated in an intraperitoneal (i.p.) tumor model, where 0.3 cm melanoma nodules develop within 3 weeks after the i.p. administration of M-21 cells. Flow cytometric and immunohistochemical analysis revealed the GD2 antigen present throughout the tumor nodule. Intraperitoneal administration of 0.01, 0.1, or 1.0 mg of ch14.18 to SCID mice previously engrafted i.p. with M-21 cells resulted in detectable ch14.18 binding to tumor cells in vivo within 10 hours of antibody administration. Ch14.18 penetration was limited to approximately 20 cell layers, demonstrating that ch14.18 has limited access to some cells in large tumor nodules. This study demonstrates that the addition of ch14.18 to IL-2 and human effector cells can result in significant antitumor activity by preventing the establishment of tumor nodules. These results suggest that clinical testing of IL-2 plus ch14.18 might be most effective if used in the setting of microscopic residual disease. Therapies that enhance ch14.18 penetration into tumor nodules should be evaluated with ch14.18 for patients with advanced melanoma.


JCO Precision Oncology | 2018

Phase I Trial of Dabrafenib and Pazopanib in BRAF Mutated Advanced Malignancies

Sigurdis Haraldsdottir; Filip Janku; Ming Poi; Cynthia Timmers; Susan Geyer; Larry J. Schaaf; Jennifer Sexton; Lai Wei; Jennifer Thurmond; Vivianne Velez-Bravo; Vanda M. Stepanek; Erin M. Bertino; Kari Kendra; Amir Mortazavi; Vivek Subbiah; Mitch A. Phelps; Manisha H. Shah

PurposeSeveral tumor types carry BRAF mutations and vascular endothelial growth factor pathway upregulation. Resistance mechanisms to BRAF inhibitors can include platelet-derived growth factor-β upregulation. Dabrafenib, a BRAF inhibitor, and pazopanib, a multikinase inhibitor that targets vascular endothelial growth factor and platelet-derived growth factor, have not been combined previously. This phase I study was designed to evaluate the safety, pharmacokinetics, and pharmacodynamics of the combination.Patients and MethodsPatients with any advanced BRAF mutated malignancy with adequate organ function were eligible. Prior use of dabrafenib or pazopanib was not allowed. Dosages started at dabrafenib 50 mg twice a day and pazopanib 400 mg daily on dose level (DL) 1, with maximum dosages of 150 mg twice a day and 800 mg daily on DL5. Pharmacokinetics and BRAF V600E plasma clone were measured, and efficacy was evaluated by imaging and tumor markers every 8 weeks.ResultsTwenty-three patients with 11 differen...


Journal of Clinical Oncology | 2017

A pilot study of interferon-alpha-2b dose reduction in melanoma: High dose interferon is not necessary for optimal activation of immune signal transduction.

Kala M. Levine; Sara Martin del Campo; Taylor R. Brooks; Elizabeth Streaker; Leslee Sprague; Volodymr Karpa; Joseph Markowitz; Anissa Bingman; Susan Geyer; Thomas Olencki; Kari Kendra; William E. Carson


Journal of Clinical Oncology | 2017

Effect of small inhibitors of nuclear export (SINE) on growth inhibition and apoptosis of human melanoma cells.

Gregory B. Lesinski; Jennifer Yang; Matthew A. Bill; Yosef Landesman; Sharon Shacham; Michael Kauffman; William Senapedis; Trinayan Kashyap; Jean-Richard Saint-Martin; Kari Kendra

Collaboration


Dive into the Kari Kendra's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jacek Gan

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jacquelyn A. Hank

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jean E. Surfus

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Lai Wei

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Paul M. Sondel

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge