Network


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

Hotspot


Dive into the research topics where Brendan D. Curti is active.

Publication


Featured researches published by Brendan D. Curti.


Journal of Clinical Oncology | 2015

Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma

Robert Hans Ingemar Andtbacka; Howard L. Kaufman; Frances A. Collichio; Thomas Amatruda; Neil Senzer; Jason Chesney; Keith A. Delman; Lynn E. Spitler; Igor Puzanov; Sanjiv S. Agarwala; Mohammed M. Milhem; Lee D. Cranmer; Brendan D. Curti; Karl D. Lewis; Merrick I. Ross; Troy H. Guthrie; Gerald P. Linette; Gregory A. Daniels; Kevin J. Harrington; Mark R. Middleton; Wilson H. Miller; Jonathan S. Zager; Yining Ye; Bin Yao; Ai Li; Susan Doleman; Ari M. Vanderwalde; Jennifer Gansert; Robert Coffin

PURPOSE Talimogene laherparepvec (T-VEC) is a herpes simplex virus type 1-derived oncolytic immunotherapy designed to selectively replicate within tumors and produce granulocyte macrophage colony-stimulating factor (GM-CSF) to enhance systemic antitumor immune responses. T-VEC was compared with GM-CSF in patients with unresected stage IIIB to IV melanoma in a randomized open-label phase III trial. PATIENTS AND METHODS Patients with injectable melanoma that was not surgically resectable were randomly assigned at a two-to-one ratio to intralesional T-VEC or subcutaneous GM-CSF. The primary end point was durable response rate (DRR; objective response lasting continuously ≥ 6 months) per independent assessment. Key secondary end points included overall survival (OS) and overall response rate. RESULTS Among 436 patients randomly assigned, DRR was significantly higher with T-VEC (16.3%; 95% CI, 12.1% to 20.5%) than GM-CSF (2.1%; 95% CI, 0% to 4.5%]; odds ratio, 8.9; P < .001). Overall response rate was also higher in the T-VEC arm (26.4%; 95% CI, 21.4% to 31.5% v 5.7%; 95% CI, 1.9% to 9.5%). Median OS was 23.3 months (95% CI, 19.5 to 29.6 months) with T-VEC and 18.9 months (95% CI, 16.0 to 23.7 months) with GM-CSF (hazard ratio, 0.79; 95% CI, 0.62 to 1.00; P = .051). T-VEC efficacy was most pronounced in patients with stage IIIB, IIIC, or IVM1a disease and in patients with treatment-naive disease. The most common adverse events (AEs) with T-VEC were fatigue, chills, and pyrexia. The only grade 3 or 4 AE occurring in ≥ 2% of T-VEC-treated patients was cellulitis (2.1%). No fatal treatment-related AEs occurred. CONCLUSION T-VEC is the first oncolytic immunotherapy to demonstrate therapeutic benefit against melanoma in a phase III clinical trial. T-VEC was well tolerated and resulted in a higher DRR (P < .001) and longer median OS (P = .051), particularly in untreated patients or those with stage IIIB, IIIC, or IVM1a disease. T-VEC represents a novel potential therapy for patients with metastatic melanoma.


The New England Journal of Medicine | 2011

gp100 Peptide Vaccine and Interleukin-2 in Patients with Advanced Melanoma

Douglas J. Schwartzentruber; David H. Lawson; Jon Richards; Robert M. Conry; Donald M. Miller; Jonathan Treisman; Fawaz Gailani; Lee B. Riley; Kevin C. Conlon; Barbara A. Pockaj; Kari Kendra; Richard L. White; Rene Gonzalez; Timothy M. Kuzel; Brendan D. Curti; Phillip D. Leming; Eric D. Whitman; Jai Balkissoon; Douglas S. Reintgen; Howard L. Kaufman; Francesco M. Marincola; Maria J. Merino; Steven A. Rosenberg; Peter L. Choyke; Don Vena; Patrick Hwu

BACKGROUND Stimulating an immune response against cancer with the use of vaccines remains a challenge. We hypothesized that combining a melanoma vaccine with interleukin-2, an immune activating agent, could improve outcomes. In a previous phase 2 study, patients with metastatic melanoma receiving high-dose interleukin-2 plus the gp100:209-217(210M) peptide vaccine had a higher rate of response than the rate that is expected among patients who are treated with interleukin-2 alone. METHODS We conducted a randomized, phase 3 trial involving 185 patients at 21 centers. Eligibility criteria included stage IV or locally advanced stage III cutaneous melanoma, expression of HLA*A0201, an absence of brain metastases, and suitability for high-dose interleukin-2 therapy. Patients were randomly assigned to receive interleukin-2 alone (720,000 IU per kilogram of body weight per dose) or gp100:209-217(210M) plus incomplete Freunds adjuvant (Montanide ISA-51) once per cycle, followed by interleukin-2. The primary end point was clinical response. Secondary end points included toxic effects and progression-free survival. RESULTS The treatment groups were well balanced with respect to baseline characteristics and received a similar amount of interleukin-2 per cycle. The toxic effects were consistent with those expected with interleukin-2 therapy. The vaccine-interleukin-2 group, as compared with the interleukin-2-only group, had a significant improvement in centrally verified overall clinical response (16% vs. 6%, P=0.03), as well as longer progression-free survival (2.2 months; 95% confidence interval [CI], 1.7 to 3.9 vs. 1.6 months; 95% CI, 1.5 to 1.8; P=0.008). The median overall survival was also longer in the vaccine-interleukin-2 group than in the interleukin-2-only group (17.8 months; 95% CI, 11.9 to 25.8 vs. 11.1 months; 95% CI, 8.7 to 16.3; P=0.06). CONCLUSIONS In patients with advanced melanoma, the response rate was higher and progression-free survival longer with vaccine and interleukin-2 than with interleukin-2 alone. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT00019682.).


Cancer | 2010

Safety and efficacy results of the advanced renal cell carcinoma sorafenib expanded access program in North America

Walter M. Stadler; Robert A. Figlin; David F. McDermott; Janice P. Dutcher; Jennifer J. Knox; Wilson H. Miller; John D. Hainsworth; Charles A. Henderson; Jeffrey R. George; Julio Hajdenberg; Tamila L. Kindwall-Keller; Marc S. Ernstoff; Harry A. Drabkin; Brendan D. Curti; Luis Chu; Christopher W. Ryan; Sebastien J. Hotte; Chenghua Xia; Lisa Cupit; Ronald M. Bukowski

The Advanced Renal Cell Carcinoma Sorafenib (ARCCS) program made sorafenib available to patients with advanced renal cell carcinoma (RCC) before regulatory approval.


Cancer Research | 2013

OX40 Is a Potent Immune-Stimulating Target in Late-Stage Cancer Patients

Brendan D. Curti; Magdalena Kovacsovics-Bankowski; Nicholas P. Morris; Edwin B. Walker; Lana Chisholm; Kevin Floyd; Joshua M. Walker; Iliana Gonzalez; Tanisha Meeuwsen; Bernard A. Fox; Tarsem Moudgil; W.R. Miller; Daniel Haley; Todd Coffey; Brenda Fisher; Laurie Delanty-Miller; Nicole Rymarchyk; Tracy L Kelly; Todd Crocenzi; Eric D. Bernstein; Rachel Sanborn; Walter J. Urba; Andrew D. Weinberg

OX40 is a potent costimulatory receptor that can potentiate T-cell receptor signaling on the surface of T lymphocytes, leading to their activation by a specifically recognized antigen. In particular, OX40 engagement by ligands present on dendritic cells dramatically increases the proliferation, effector function, and survival of T cells. Preclinical studies have shown that OX40 agonists increase antitumor immunity and improve tumor-free survival. In this study, we performed a phase I clinical trial using a mouse monoclonal antibody (mAb) that agonizes human OX40 signaling in patients with advanced cancer. Patients treated with one course of the anti-OX40 mAb showed an acceptable toxicity profile and regression of at least one metastatic lesion in 12 of 30 patients. Mechanistically, this treatment increased T and B cell responses to reporter antigen immunizations, led to preferential upregulation of OX40 on CD4(+) FoxP3(+) regulatory T cells in tumor-infiltrating lymphocytes, and increased the antitumor reactivity of T and B cells in patients with melanoma. Our findings clinically validate OX40 as a potent immune-stimulating target for treatment in patients with cancer, providing a generalizable tool to favorably influence the antitumor properties of circulating T cells, B cells, and intratumoral regulatory T cells.


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 Clinical Oncology | 1992

The toxic and hematologic effects of interleukin-1 alpha administered in a phase I trial to patients with advanced malignancies.

J W Smith nd; Walter J. Urba; Brendan D. Curti; L J Elwood; Ronald G. Steis; John E. Janik; William H. Sharfman; L L Miller; Robert G. Fenton; Kevin C. Conlon

PURPOSE A phase I trial was undertaken because interleukin-1 alpha (IL-1 alpha) possesses antiproliferative, immunostimulatory, antiinfection, myeloprotective, and myelorestorative properties that could be beneficial in cancer treatment. PATIENTS AND METHODS In this phase I trial, IL-1 alpha was administered intravenously (IV) during a 15-minute period daily for 7 days to patients with advanced solid malignancies. RESULTS The maximum-tolerated dose (MTD) of IL-1 alpha alone was 0.3 microgram/kg. A second group of patients received indomethacin plus IL-1 alpha based on preclinical studies, which indicated that indomethacin could abrogate IL-1 alpha-induced hypotension; however, the MTD of IL-1 alpha plus indomethacin was 0.1 microgram/kg lower than IL-1 alpha alone. Fever, chills, headache, nausea, vomiting, and myalgia were common but were not dose-limiting. Hypotension resulted from a marked decrease in systemic vascular resistance and required pressors at 0.3 and 1.0 micrograms/kg IL-1 alpha. Dose-limiting toxicities included hypotension, myocardial infarction, confusion, severe abdominal pain, and renal insufficiency. IL-1 alpha treatment caused a significant, dose-related increase in the total WBC count (mainly segmented neutrophils and neutrophilic bands). Bone marrow cellularity increased because of enhanced numbers of relatively mature myeloid cells and megakaryocytes. Platelet counts decreased during therapy but were significantly elevated above baseline values 1 to 2 weeks posttreatment; this may have been an effect of IL-6 that was shown to be induced by IL-1 alpha treatment. Significant increases in triglycerides, cortisol, C-reactive protein, thyroid-stimulating hormone and decreases in cholesterol, testosterone, and protein-C were observed with treatment. CONCLUSION We conclude that at doses of IL-1 alpha that can be given safely to cancer patients, significant, potentially beneficial hematopoietic effects occur.


Journal of Clinical Oncology | 2007

Phase I/II Trial of Temsirolimus Combined With Interferon Alfa for Advanced Renal Cell Carcinoma

Robert J. Motzer; Gary R. Hudes; Brendan D. Curti; David F. McDermott; Bernard Escudier; Sylvie Négrier; Brigitte Duclos; Laurence Moore; Timothy O'Toole; Joseph Boni; Janice P. Dutcher

PURPOSE Temsirolimus, an inhibitor of the mammalian target of rapamycin, has single-agent activity against advanced renal cell carcinoma (RCC). A recommended dose and safety profile for the combination of temsirolimus and interferon alfa (IFN) were determined in patients with advanced RCC. PATIENTS AND METHODS Patients were enrolled onto a multicenter, ascending-dose study of temsirolimus (5, 10, 15, 20, or 25 mg) administered intravenously once a week combined with IFN (6 or 9 million units [MU]) administered subcutaneously three times per week. An expanded cohort was treated at the recommended dose to obtain additional safety and efficacy information. RESULTS Seventy-one patients were entered to receive one of six dose levels. The recommended dose was temsirolimus 15 mg/IFN 6 MU based on dose-limiting toxicities of stomatitis, fatigue, and nausea/vomiting, which were observed at higher doses of temsirolimus and IFN. The most frequent grade 3 or 4 toxicities occurring in any cycle included leukopenia, hypophosphatemia, asthenia, anemia, and hypertriglyceridemia for all patients and those who received the recommended dose. Among patients who received the recommended dose (n = 39), 8% achieved partial response and 36% had stable disease for at least 24 weeks. Median progression-free survival for all patients in the study was 9.1 months. CONCLUSION The combination of temsirolimus and IFN has an acceptable safety profile and displays antitumor activity in patients with advanced RCC. Temsirolimus 15 mg plus IFN 6 MU is the recommended dose for evaluation in a randomized phase III study.


Nature Reviews Clinical Oncology | 2013

The Society for Immunotherapy of Cancer consensus statement on tumour immunotherapy for the treatment of cutaneous melanoma

Howard L. Kaufman; John M. Kirkwood; F. Stephen Hodi; Sanjiv S. Agarwala; Thomas Amatruda; Steven D. Bines; Joseph I. Clark; Brendan D. Curti; Marc S. Ernstoff; Thomas F. Gajewski; Rene Gonzalez; Laura Jane Hyde; David H. Lawson; Michael T. Lotze; Jose Lutzky; Kim Margolin; David F. McDermott; Donald L. Morton; Anna C. Pavlick; Jon Richards; William H. Sharfman; Vernon K. Sondak; Jeffrey A. Sosman; Susan Steel; Ahmad A. Tarhini; John A. Thompson; Jill Titze; Walter J. Urba; Richard L. White; Michael B. Atkins

Immunotherapy is associated with durable clinical benefit in patients with melanoma. The goal of this article is to provide evidence-based consensus recommendations for the use of immunotherapy in the clinical management of patients with high-risk and advanced-stage melanoma in the USA. To achieve this goal, the Society for Immunotherapy of Cancer sponsored a panel of melanoma experts—including physicians, nurses, and patient advocates—to develop a consensus for the clinical application of tumour immunotherapy for patients with melanoma. The Institute of Medicine clinical practice guidelines were used as a basis for this consensus development. A systematic literature search was performed for high-impact studies in English between 1992 and 2012 and was supplemented as appropriate by the panel. This consensus report focuses on issues related to patient selection, toxicity management, clinical end points and sequencing or combination of therapy. The literature review and consensus panel voting and discussion were used to generate recommendations for the use of immunotherapy in patients with melanoma, and to assess and rate the strength of the supporting evidence. From the peer-reviewed literature the consensus panel identified a role for interferon-α2b, pegylated-interferon-α2b, interleukin-2 (IL-2) and ipilimumab in the clinical management of melanoma. Expert recommendations for how to incorporate these agents into the therapeutic approach to melanoma are provided in this consensus statement. Tumour immunotherapy is a useful therapeutic strategy in the management of patients with melanoma and evidence-based consensus recommendations for clinical integration are provided and will be updated as warranted.


The New England Journal of Medicine | 1993

The effects of treatment with interleukin-1α on platelet recovery after high-dose carboplatin

John W. Smith; Dan L. Longo; W. Gregory Alvord; John E. Janik; William H. Sharfman; Barry L. Gause; Brendan D. Curti; Stephen P. Creekmore; Jon T. Holmlund; Robert G. Fenton; Mario Sznol; Langdon L. Miller; Masanao Shimizu; Joost J. Oppenheim; Shelby J. Fiem; Jean Hursey; Gerry C. Powers; Walter J. Urba

Background Thrombocytopenia is a frequent side effect of cancer chemotherapy and commonly limits attempts to escalate drug doses. To determine whether interleukin-1α could ameliorate carboplatin-induced thrombocytopenia, we combined it with high-dose carboplatin in 43 patients with advanced neoplasms. Methods High-dose carboplatin (800 mg per square meter of body-surface area) was administered alone to a control group. Subsequent patients were randomly assigned to receive the same dose of carboplatin with interleukin-1α, administered either before or after carboplatin. Interleukin-1α was given intravenously at a dose of 0.03, 0.1, or 0.3 μg per kilogram of body weight per day for five days. Results Carboplatin alone consistently produced thrombocytopenia with a median nadir of 19,000 platelets per cubic millimeter and a median of 10 days with less than 100,000 platelets per cubic millimeter. All 15 patients receiving interleukin-1α before carboplatin had similar findings. In contrast, 5 of the 15 patients...


Immunological Reviews | 2011

Science gone translational: the OX40 agonist story

Andrew D. Weinberg; Nicholas P. Morris; Magdalena Kovacsovics-Bankowski; Walter J. Urba; Brendan D. Curti

Summary:  OX40 (CD134) is a tumor necrosis factor (TNF) receptor expressed primarily on activated CD4+ and CD8+ T cells and transmits a potent costimulatory signal when engaged. OX40 is transiently expressed after T‐cell receptor engagement and is upregulated on the most recently antigen‐activated T cells within inflammatory lesions (e.g. sites of autoimmune destruction and on tumor‐infiltrating lymphocytes). Hence, it is an attractive target to modulate immune responses: OX40 blocking agents to inhibit undesirable inflammation or OX40 agonists to enhance immune responses. In regards to this review, OX40 agonists enhance anti‐tumor immunity, which leads to therapeutic effects in mouse tumor models. A team of laboratory and clinical scientists at the Providence Cancer Center has collaborated to bring the preclinical observations in cancer models from the bench to the bedside. This review describes the journey from in vitro experiments through preclinical mouse models to the successful translation of the first OX40 agonist to the clinic for the treatment of patients with cancer.

Collaboration


Dive into the Brendan D. Curti's collaboration.

Top Co-Authors

Avatar

Walter J. Urba

Providence Portland Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dan L. Longo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David F. McDermott

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joseph I. Clark

Loyola University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Janice P. Dutcher

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John E. Janik

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge